ArticlePDF AvailableLiterature Review

Abstract and Figures

The protein encoded by the TP53 gene is one of the most important suppressors of tumor formation, which is also frequently inactivated in gastrointestinal cancer. MicroRNAs (miRNAs) are small noncoding RNAs that inhibit translation and/or promote degradation of their target messenger RNAs. In recent years, several miRNAs have been identified as mediators and regulators of p53's tumor suppressing functions. p53 induces expression and/or maturation of several miRNAs, which leads to the repression of critical effector proteins. Furthermore, certain miRNAs regulate the expression and activity of p53 through direct repression of p53 or its regulators. Experimental findings indicate that miRNAs are important components of the p53 network. In addition, the frequent genetic and epigenetic alterations of p53-regulated miRNAs in tumors indicate that they play an important role in cancer initiation and/or progression. Therefore, p53-regulated miRNAs may represent attractive diagnostic and/or prognostic biomarkers. Moreover, restoration of p53-induced miRNAs results in suppression of tumor growth and metastasis in mouse models of cancer. Thus, miRNA-based therapeutics may represent a feasible strategy for future cancer treatment. Here we summarize the current published state-of-the-art on the role of the p53-miRNA connection in gastrointestinal cancer.
This content is subject to copyright. Terms and conditions apply.
© 2014 Rokavec et al. This work is published by Dove Medical Press Limited, and licensed under Creative Commons Attribution – Non Commercial (unported, v3.0)
License. The full terms of the License are available at http://creativecommons.org/licenses/by-nc/3.0/. Non-commercial uses of the work are permitted without any further
permission from Dove Medical Press Limited, provided the work is properly attributed. Permissions beyond the scope of the License are administered by Dove Medical Press Limited. Information on
how to request permission may be found at: http://www.dovepress.com/permissions.php
Clinical and Experimental Gastroenterology 2014:7 395–413
Clinical and Experimental Gastroenterology Dovepress
submit your manuscript | www.dovepress.com
Dovepress 395
REVIEW
open access to scientific and medical research
Open Access Full Text Article
http://dx.doi.org/10.2147/CEG.S43738
The p53/microRNA connection
in gastrointestinal cancer
Matjaz Rokavec
Huihui Li
Longchang Jiang
Heiko Hermeking
Experimental and Molecular
Pathology, Institute of Pathology,
Ludwig-Maximilians-Universität
München, Munich, Germany
Correspondence: Heiko Hermeking
Experimental and Molecular Pathology,
Institute of Pathology,
Ludwig-Maximilians-University Munich,
Thalkirchner Strasse 36,
D-80337 Munich, Germany
Tel +49 89 2180 73 685
Fax +49 89 2180 73 697
Email heiko.hermeking@med.uni-
muenchen.de
Abstract: The protein encoded by the TP53 gene is one of the most important suppressors of
tumor formation, which is also frequently inactivated in gastrointestinal cancer. MicroRNAs
(miRNAs) are small noncoding RNAs that inhibit translation and/or promote degradation
of their target messenger RNAs. In recent years, several miRNAs have been identified as
mediators and regulators of p53’s tumor suppressing functions. p53 induces expression and/
or maturation of several miRNAs, which leads to the repression of critical effector proteins.
Furthermore, certain miRNAs regulate the expression and activity of p53 through direct
repression of p53 or its regulators. Experimental findings indicate that miRNAs are important
components of the p53 network. In addition, the frequent genetic and epigenetic alterations of
p53-regulated miRNAs in tumors indicate that they play an important role in cancer initiation
and/or progression. Therefore, p53-regulated miRNAs may represent attractive diagnostic and/
or prognostic biomarkers. Moreover, restoration of p53-induced miRNAs results in suppression
of tumor growth and metastasis in mouse models of cancer. Thus, miRNA-based therapeutics
may represent a feasible strategy for future cancer treatment. Here we summarize the current
published state-of-the-art on the role of the p53-miRNA connection in gastrointestinal cancer.
Keywords: p53, microRNA, cancer, gastrointestinal tract
Introduction
Gastrointestinal (GI) cancers represent malignant tumors of the GI tract and accessory
organs of digestion. They include carcinomas arising in the oral cavity, esophagus,
stomach, liver, gallbladder, pancreas, small intestine, large intestine, rectum, and anus.
GI cancer represents about 30% of all tumor incidences and is responsible for approxi-
mately 40% of tumor-related mortality worldwide (Figure 1A and B).1 Tumors of the
GI tract harbor mutations in the p53 tumor suppressor gene (TP53) with a prevalence
ranging from 31% to 45% (Figure 1C).2 The p53 protein functions as a transcription
factor that mediates the response to many cellular stresses, most prominently the
DNA damage response. p53 suppresses a variety of malignant processes, thereby
representing one of the most important cancer suppressing proteins.3 p53 protects
against cancer by inducing cellular processes such as apoptosis or cell cycle arrest,
thereby preventing the propagation of damaged cells that potentially could give rise to
tumors.4,5 Moreover, p53 inhibits epithelial to mesenchymal transition (EMT), stem-
ness, and metabolic adaptations, which are typically found in tumors.6 In addition,
p53 promotes DNA repair, antioxidant defense, and differentiation. On the molecular
level, p53 exerts its tumor suppressive functions by regulating the expression of numer-
ous target genes, mainly by direct binding to specific DNA motifs located in target
Number of times this article has been viewed
This article was published in the following Dove Press journal:
Clinical and Experimental Gastroenterology
30 September 2014
Clinical and Experimental Gastroenterology 2014:7
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
396
Rokavec et al
gene promoters.7 Besides p53-regulated protein expression,
p53-induced microRNAs (miRNAs) have emerged as impor-
tant effectors of p53.8,9 The generation of mature miRNAs
is a multistage process (see Figure 2) starting with the tran-
scription of miRNA encoding genes to yield the primary
miRNA (pri-miRNA).10 Next, the pri-miRNA is cleaved by
the RNAse III enzyme Drosha, resulting in a 70 nucleotide
stem-loop-structured miRNA precursor molecule (pre-
miRNA).11 The pre-miRNA is transported to the cytoplasm
by Exportin 5, where it is cleaved further by the RNAse
Dicer. The resulting 20 to 25 bp mature miRNA is incorpo-
rated into the miRNA-induced silencing complex (miRISC),
which mediates miRNA-induced silencing of target mes-
senger RNAs (mRNAs).12 miRNAs bind to 3-untranslated
regions (3-UTR) of mRNAs via their seed sequences, which
are conserved seven nucleotide regions in their 5 region. The
association of the miRISC with seed-matching sequences
in target mRNAs results in the inhibition of translation and
degradation of the target mRNAs.10 It has been estimated
that .60% of human protein coding genes are subject to
regulation by miRNAs.13 Not surprisingly, miRNA-mediated
regulation has been implicated in almost all physiological
and pathophysiological processes.10 Interestingly, several
miRNAs may also be of use for diagnostic, prognostic,
and therapeutic applications in GI-cancers.14–18 Extracel-
lular miRNAs have been detected in blood serum. These
miRNAs are either secreted by living cells via exosomes or
microvesicles, or they originate from dying cells.19 Interest-
ingly, these circulating miRNAs are extremely stable, both in
blood and after isolation. Numerous studies have shown their
potential usefulness as noninvasive diagnostic and prognos-
tic markers in GI cancers.20 Seven years ago it was shown
that p53 also regulates the expression of miRNA-encoding
genes.8 The p53-regulated miRNAs have been implicated
Figure 1 Incidence (A) and mortality (B) of indicated gastrointestinal (GI) cancers worldwide. (C) Prevalence of mutations in the TP53 gene in the indicated GI cancers.
Clinical and Experimental Gastroenterology 2014:7 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
397
The p53/microRNA axis in GI cancer
in the control of various cancer-related processes, such
as proliferation, apoptosis, EMT, migration, invasion, and
metastasis. Therefore, they may represent important media-
tors of the tumor suppressive function of p53. In addition,
a number of miRNAs can regulate expression and activity
of the p53 protein, either negatively through direct repres-
sion of p53 expression, or positively through the repression
of negative regulators of p53. In this review we summarize
the current knowledge about the p53/miRNA network and
its role in GI cancers.
Figure 2 Effects of p53 on miRNA biogenesis.
Notes: Wild-type p53 (green) regulates miRNA transcription, processing, and target selection. In contrast, mutant p53 (blue) is unable to induce the expression of miRNAs
and additionally inhibits the p63-mediated activation of the miRNA processing protein Dicer.
Abbreviations: mRNA, messenger RNA; miRNA, microRNA; pre-microRNA, miRNA precursor molecule; pri-microRNA, primary miRNA; miRISC, miRNA-induced
silencing complex; mut, mutant; RE, response element.
Clinical and Experimental Gastroenterology 2014:7
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
398
Rokavec et al
Table 1 Summary of changes in expression of p53-pathway-related miRNAs in GI cancers
Notes: For a more detailed description of the single miRNAs and references see Tables 2 and 3. Green box with : downregulated in indicated GI tumor type; Red box
with ∆: upregulated in indicated GI tumor type; Yellow box with /∆: down- or upregulated in indicated GI tumors.
Abbreviations: CRC, colorectal cancer; EC, esophageal cancer; GC, gastric cancer; GI, gastrointestinal; HCC, hepatocellular cancer; miRNA, microRNA; PaC, pancreatic cancer.
p53 regulated miRNAs
In 2007, we and other groups identified several miRNAs as
direct transcriptional targets of p53.21–27 Since then, many of
these miRNAs have been validated as important mediators of
p53 functions (Table 1 and 2).9 p53 regulates the expression
of its target miRNAs either on the transcriptional level by
direct binding to the promoters of the corresponding genes,
or by regulating miRNA processing (Figure 2). It was shown
that p53 interacts with the DEAD-box RNA helicase p68
(also known as DDX5) and enhances its interaction with the
Drosha complex. As a result, p53 promotes the processing of
specific pri-miRNAs to pre-miRNAs, leading to elevated lev-
els of the respective miRNAs.28 Another link between p53 and
miRNA-processing has been observed in conditional Dicer
knockout mice.29 Dicer deficiency, and therefore incomplete
miRNA maturation, induces p53, which leads to
Clinical and Experimental Gastroenterology 2014:7 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
399
The p53/microRNA axis in GI cancer
Table 2 Compilation of p53-regulated miRNAs and their alterations in GI cancers
miRNA Validated by Clinical and pathological associations in GI cancers
miRNAs induced by p53
miR-34a Luc reporter (mut), qPCR, ChIP24 CRC: downregulated in tumors38 and serum;162 Downregulation in metastatic tumors;70,163 CpG
methylation66,67 in metastatic tumors70
EC, HCC: downregulated in tumors;39,41 CpG methylation39,41,164
GC: downregulated in tumors40
PaC: downregulation associated with poor OS;165 CpG methylation66,67
miR-34b/c Luc reporter (mut), qPCR, ChIP24 CRC, GC, PaC: CpG methylation67,69
EC: upregulation associated with advanced tumor stage;166 CpG methylation164
HCC: downregulated in tumors41
miR-15a/16-1 qPCR, ChIP33 CRC: miR-16-1 down-regulated in tumors;77 Downregulation of miR-16-1 associated with pN,
TNM stage, and poor OS77
EC: miR-15a upregulated in tumors167
miR-200c/141 Luc reporter (mut), qPCR, ChIP32 CRC: downregulation of miR-200c is associated with poor OS;168 miR-200c is downregulated
in metastatic CRC;169 miR-200c is upregulated in patient serum;112,170 Upregulation in serum is
associated with tumor stage, pN, metastasis, and prognosis;109 CpG methylation171
EC: miR-200c is upregulated in patient serum;108 Upregulation in serum associated with poor RFS108
GC: miR-141 is downregulated in tumors172
HCC: miR-200c is downregulated in tumors;173 Downregulation associated with poor RFS174
miR-200a/
200b/429
Luc reporter (mut), qPCR, ChIP32 CRC: miR-429 is downregulated in tumors175; downregulation of miR-200a/429 is associated
with poor OS;168 CpG methylation171
EC: miR-200a/b are upregulated in tumors167,176
GC: miR-200a is downregulated in tumors176
PaC: CpG hypomethylation and overexpression in tumors177
miR-107 Luc reporter (mut), qPCR, ChIP36 CRC: upregulation associated with metastasis and poor OS117
EC: downregulated in tumors and serum178
GC: upregulated in tumors; Upregulation associated with tumor invasion, stage, pN, and poor
DFS and OS119
PaC: CpG methylation179
miR-145 Luc reporter (mut), qPCR, ChIP35 CRC: downregulated in tumors180,181 and patient stool samples;182 Downregulation associated
with tumor size91
EC, GC, HCC, PaC: downregulated in tumors88–90,173,183,184
miR-192/
194/215
qPCR, ChIP34 CRC: downregulation of miR-192/194/215 in tumors;34,185,186 Downregulation of miR-192/215
associated with stage, grade, pN;103,186 Downregulation of miR-215 associated with poor RFS;101
Upregulation of miR-194 associated with poor RFS and OS187
GC: downregulation of miR-194 associated with higher tumor size and stage;188 miR-215
upregulated in tumors189
miR-29 Luc reporter, qPCR149 CRC: upregulated in serum;190 Upregulation associated with metastasis and OS191
GC: downregulated in tumors; Downregulation associated with metastasis152,192
HCC: downregulated in tumors; Downregulation associated with poor RFS and OS153,154
miR-605 Luc reporter (mut), qPCR, ChIP193 NA
miR-149 Luc reporter (mut), qPCR, ChIP194 CRC: downregulated in tumors;195 CpG methylation;196 Downregulation associated with
invasion and poor OS196
GC: downregulated in tumors197
miR-22 Luc reporter (mut), qPCR, ChIP198 CRC: downregulated in tumors and liver metastases; Downregulation associated with poor OS199
EC, PaC: upregulated in serum200,201
GC: downregulated in tumors;202 Downregulation associated with tumor stage, pN, metastasis,
poor OS203
HCC: downregulated in tumors, Downregulation associated with pN, grade, and poor OS204,205
miR-23b Luc reporter (mut), ChIP206 EC: downregulation associated with poor prognosis207
CRC, HCC: downregulated in tumors208,209
miR-205 Luc reporter (mut), qPCR, ChIP210 EC: downregulated in tumors,211,212 Down-regulation associated with poor prognosis207
HCC: downregulated in tumors213
miR-1246 Luc reporter (mut), qPCR, ChIP214 CRC: downregulated in tumors215; Upregulated in patient serum216
EC: upregulated in tumors217 and patient serum218
miR-1204 qPCR, ChIP219 NA
(Continued)
Clinical and Experimental Gastroenterology 2014:7
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
400
Rokavec et al
reduced proliferation and premature senescence. Therefore,
p53 may operate as a checkpoint to monitor proper miRNA
processing. Moreover, expression of Dicer1 is regulated by the
p53 family member p63, which can be inhibited by associa-
tion with mutant p53.30 Finally, p53 also affects miRNA target
gene selection by regulating mRNA-binding proteins, such as
RNA-binding-motif protein 38, which competes with miR-
NAs for binding to 3-UTRs of mRNAs.31 miRNAs transcrip-
tionally induced by p53 include the miR-34,21–26 miR-200,32
miR-15a/16-1,33 and miR-192/194/21534 clusters, as well as
miR-14535 and miR-107.36 Yet some of these miRNAs, such as
miR-16-1, miR-145, and miR- 199a-3p, are also regulated on
the post-transcriptional level by p53.28,37 The expression of the
majority of these miRNAs is frequently altered in GI tumors
and has been associated with clinical and pathological param-
eters of various types of GI cancer (Tables 1 and 2).
The miR-34 family
The miR-34 family includes three members miR-34a,
miR-34b, and miR-34c which show a marked induc-
tion by p53.8 MiR-34a is encoded by its own host gene,
whereas miR-34b and miR-34c share a common precursor.
Both miR-34 genes contain several p53-responsive
elements, which are occupied by p53 and mediate activa-
tion of miR- 34a/b/c after DNA damage.25,26 Expression of
miR-34a/b/c is frequently downregulated in colorectal,38
esophageal,39 gastric,40 and hepatocellular cancers (HCCs).41
Consistently, all members of the miR-34 family were shown
to suppress tumor growth and metastasis by inhibiting pro-
cesses that promote cancer, including cell cycle progression,
EMT, metastasis, and stemness and by promoting tumor
suppressive processes, such as apoptosis and senescence.42
MiR-34s regulate these processes through suppressing the
expression of their target mRNAs, such as SNAIL, c-Myc,
Bcl2, c-Met, and Axl.43 The miR-34/p53 axis and its targets
are often connected through positive or negative feedback
loops that either reinforce the miR-34/p53 signaling or
suppress it. For example, a positive feedback loop connects
miR-34a and p53 via MDM4 (Figure 3A). MDM4 and
its human counterpart HDM4 bind to p53 and inhibit its
transcriptional activity. At the same time MDM4/HDM4
are targets of miR-34a.44,45 Therefore, the repression of
MDM4/HDM4 by miR-34a leads to stabilization of p53 and
enhanced expression of miR-34a. Recently, it was shown that
in addition to full-length HDM4 that is targeted by miR-34a,
a short isoform of HDM4 also exists, which lacks seed-
matching sites for miR-34a, thereby evading suppression
by miR-34a.45 Consistently, this short HDM4 isoform was
highly expressed in tumors, where it presumably inhibits
the miR-34a/p53 axis.
Furthermore, SIRT1 was shown to mediate activation
of p53 by miR-34a.46 SIRT1 is an nicotinamide adenine
dinucleotide (NAD+)-dependent deacetylase, which represses
p53 activity by deacetylation of p53 protein. Yamakuchi
et al showed that SIRT1 is a miR-34 target and that miR-34
induces the activity of p53 by repressing SIRT1 in colorectal
cancer (CRC) cell lines (Figure 3B). Moreover, miR-34 not
only represses the expression of SIRT1, but also suppresses
SIRT1 activity by downregulating nicotinamide phospho-
ribosyltransferase (NAMPT), the rate-limiting enzyme of
NAD+ biosynthesis.47 Furthermore, SIRT1 and MYC regulate
each other via a positive feedback loop.48,49 By repressing
both SIRT1 and MYC, miR-34a may therefore efficiently
suppress this circuitry.
Another double-negative feedback loop involving miR-
34a was discovered by Siemens et al,50 who demonstrated that
miR-34a directly targets and suppresses the EMT-inducing
transcription factor (EMT-TF) SNAIL, whereas SNAIL
represses the miR-34a and miR-34b/c genes by directly
binding to their promoters in CRC cell lines (Figure 3C).
By utilizing HCC and CRC cells, Kim et al showed that
p53 regulates EMT by inducing members of the miR-200
Table 2 (Continued)
miRNA Validated by Clinical and pathological associations in GI cancers
miRNAs repressed by p53
miR-17-92 Luc reporter (mut), qPCR, ChIP126 CRC,220,221 EC,222,223 GC,224,225 HCC,226 PaC;227 Upregulated in tumors
miR-224 Luc reporter (mut), qPCR, ChIP124 CRC: upregulated in tumors;228,229 Upregulation associated with poor RFS and OS;228,229
Downregulation associated with poor OS;230 Upregulation associated with tumor size, stage,
and metastasis229
HCC: upregulated in tumors231,232
miR-502 Luc reporter125 CRC: downregulated in tumors125
Abbreviations: ChIP, chromatin immunoprecipitation; CRC, colorectal cancer; DFS, disease-free survival; EC, esophageal cancer; GC, gastric cancer; GI, gastrointestinal;
HCC, hepatocellular cancer; Luc reporter, Luciferase miRNA promoter reporter assay; miRNA, microRNA; mut, mutation in the p53 binding site; NA, not
applicable or not analyzed; OS, overall survival; PaC, pancreatic cancer; pN, nodal status; qPCR, quantitative real-time polymerase chain reaction; RFS, relapse-free
survival; TNM, tumor, node, metastasis status based classication.
Clinical and Experimental Gastroenterology 2014:7 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
401
The p53/microRNA axis in GI cancer
Figure 3 The role of p53/miRNA axis in (A) p53 autoregulation, (B) cancer cell metabolism, (C) invasion, and metastasis, as a result of the regulation of EMT/MET (D)
cancer-associated inammatory signaling.
Notes: Color code: p53/green; miRNAs/yellow; miRNA-targets/red; green arrow/activation; red arrow/inhibition.
Abbreviations: EMT, epithelial–mesenchymal transition; IL-6, interleukin 6; IL-6R, interleukin 6 receptor; MET, mesenchymal–epithelial transition; miRNA, microRNA;
NAD+, nicotinamide adenine dinucleotide; NAMPT, nicotinamide phosphoribosyltransferase.
family,32 which also represent EMT-regulating miRNAs that
suppress EMT by a similar double-negative feedback loop
involving the EMT-TFs ZEB1 and ZEB2.51,52 Thus, p53 is a
key regulator of cellular plasticity by controlling EMT and
its counterpart mesenchymal–epithelial transition (MET)
through inducing the miRNAs of the miR-34 and miR-200
family (Figure 3C). These miRNAs form two double-
negative feedback loops with their targets SNAIL, ZEB1,
and ZEB2 that act as bimodal switches to stabilize either
the epithelial or the mesenchymal state. Moreover, ZEB1
Clinical and Experimental Gastroenterology 2014:7
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
402
Rokavec et al
was also shown to repress miR-34a expression by binding
to the same E-boxes in the miR-34 promoters as SNAIL,
thereby further interconnecting the miR-34/SNAIL and
miR-200/ZEB loops.50,53 In addition, we recently showed
that the zinc-finger 281 (ZNF281) protein is an important
miR-34 target with respect to EMT.54 We found that the
expression of ZNF281 is controlled by miR-34 and SNAIL
in a coherent feed-forward-loop, whereby SNAIL and
ZNF281 induce each other, whereas miR-34 can directly
repress both SNAIL and ZNF281 (Figure 3C). Accord-
ingly, ectopic SNAIL induced EMT by directly activating
ZNF281 and concomitantly repressing miR-34a expres-
sion, which leads to a further increase in ZNF281 levels.
Notably, the induction of ZNF281 by SNAIL was required
for SNAIL-induced EMT.
Recently, we demonstrated that inflammation can
suppress the expression of miR-34a.55 We showed that
exposure to the proinflammatory cytokine interleukin-6
(IL-6) results in repression of miR-34a via direct binding
of the IL-6 effector STAT3 to the first intron of the miR-
34a gene. Furthermore, we identified the IL-6 receptor
(IL-6R) as a direct target of miR-34a. Further functional
analysis revealed the existence of an IL-6R/STAT3/miR-
34a feedback loop (Figure 3D). The activation of this loop
was required for EMT, invasion, and metastasis of CRC
cell lines and was associated with nodal and distant metas-
tasis in CRC patients. Moreover, in miR-34a-deficient
mice, colitis-associated intestinal tumors displayed acti-
vation of the loop and, in contrast to tumors in wild-type
mice, progressed to invasive carcinomas. Our findings
suggest that the activation of the IL-6R/STAT3/miR-34a
loop by IL-6 drives cancer cells toward a mesenchymal
and invasive phenotype, whereas suppression of the loop
by p53 shifts cancer cells toward an epithelial state and
prevents EMT and invasion.55
Reintroduction of miR-34 into tumors, which lost
miR-34 expression, may represent an attractive alterna-
tive for cancer treatment. The most common approach for
miRNA delivery relies on lipid-based nanoparticles, which
contain vectors expressing miRNAs, or 19–23-nt double-
stranded mimics of mature miRNAs. These can be admin-
istered systemically by intravenous injection or locally into
tumors. Several studies have shown that systemic miR-34a
delivery suppresses tumor growth in vivo. Using xenograft
or genetically engineered mouse models of melanoma,
lymphoma, multiple myeloma, breast, prostate, pancreatic,
and non-small-cell lung cancer, the authors observed an
inhibition of tumor growth by 20% to 83% after reintroduc-
tion of miR-34a.56 Importantly, no severe toxicity caused by
systemic miR-34a delivery has been observed in mice.57,58
Likewise, no unwanted immune response has been detected,
based on serum cytokine levels in immune-competent mice.59
Recently, the company Mirna Therapeutics has initiated a
clinical Phase I trial of nanoparticle-based delivery of miR-
34a in patients with non-resectable primary liver cancer or
metastatic cancer with liver involvement.60 Therefore, miR-
34a may be one of the first miRNA mimics to reach the clinic.
Conventional anticancer therapies, such as chemotherapy
and treatment with radiation, induce miR-34 expression in
human cancer cells with wild-type p53.27 However, since the
majority of human tumors lack normal p53 function, replace-
ment of miR-34 may enhance the efficacy of standard cancer
therapies. Indeed, in prostate, colorectal, and bladder cancer
cells, reintroduction of miR-34a precursors enhanced the
sensitivity toward camptothecin, paclitaxel, 5-fluorouracil,
and cisplatin.61–65 Furthermore, lentiviral transduction of
miR-34a sensitized gastric and pancreatic cancer cells to
radiation and to the chemotherapeutic drugs docetaxel,
gemcitabine, cisplatin, and doxorubicin.63,64 Moreover, we
recently showed that c-Kit is an important miR-34a target
that mediates, at least in part, chemosensitization by miR-
34a in CRC cell lines.65 Thus, these results suggest that
combined treatment with miR-34 mimics may enhance the
beneficial effects of conventional cancer therapies.
Downregulation of miR-34 expression in tumors has
been frequently attributed to methylation of the CpG islands
present in promoters of miR-34a and miR-34b/c.66–69 CpG
methylation is causally involved in repression of miR-34-
a/b/c, since treatment of CRC cell lines with the demethylat-
ing agent 5-aza-2-deoxycytidine leads to re-expression of
miR-34a/b/c.66,68,69 Moreover, a significant inverse correlation
between miR-34a methylation and expression has been
observed in colon tumors.70,71 Therefore, miRNA cancer
treatment strategies may rely not only on delivery of syn-
thetic miRNA mimics corresponding to miR-34a/b/c into
tumors, but also on re-expression of these miRNAs using
demethylating agents. Indeed, treatment with BioResponse
3,3-Diindolylmethane, an experimental anti-androgen pros-
tate cancer drug, resulted in demethylation and re-expression
of miR-34a in prostate cancer cells.72 In a Phase II clinical
trial, treatment of prostate cancer patients with BioResponse
3,3-Diindolylmethane prior to radical prostatectomy led
to the re-expression of miR-34a as well as repression and
nuclear exclusion of its target, the androgen receptor.72,73
Several reports showed that miR-34 methylation may also
have prognostic value. In our study, miR-34a methylation
Clinical and Experimental Gastroenterology 2014:7 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
403
The p53/microRNA axis in GI cancer
in primary CRC was significantly associated with increased
formation of lymph node and liver metastases.70 Recently,
Wu et al analyzed miR-34a/b/c methylation in stool samples
of 82 CRC patients and 40 controls.74 They demonstrated
that detection of miR-34a and miR-34b/c methylation could
identify CRC with a remarkable sensitivity of 76.8% or 95%
and a specificity of 93.6% or 100%, respectively. Therefore,
the detection of miR-34 miRNAs and CpG methylation of
miR-34 promoter regions in body fluids or stool represent
potential biomarkers which may be utilized for noninvasive
screening and diagnosis of cancer in the future.
miR-15a and miR-16-1
Mir-15a and miR-16-1 were the first miRNAs genetically
linked to cancer: In 2002, Calin et al showed that miR-15a and
miR-16-1, which are encoded within an intron of the DLEU2
gene, are frequently deleted and/or downregulated in chronic
lymphocytic leukemia.75 Notably, a knockout of DLEU2 or of
the miR-15a/16-1-bearing intron in mice confirmed that loss
of miR-15a/16-1 causes chronic lymphocytic leukemia.76
The expression of miR-15a and miR-16-1 is induced by p53
via transcriptional33 and post-transcriptional mechanisms.28
Several studies implicated the downregulation or loss
of miR-15a and miR-16-1 expression in GI cancers. For
example, the expression of miR-16-1 was significantly
lower in primary CRC when compared to the corresponding
normal colonic mucosa.77 Moreover, decreased miR-16-1
expression was associated with lymph node metastasis and
recurrence of colorectal tumors.77 Furthermore, ectopic
expression of miR-15a and miR-16-1 inhibited the prolif-
eration of pancreatic and colorectal cancer cells78,79 and led
to a significant inhibition of subcutaneous growth of CRC
cell lines in immune-compromised mice.80 Interestingly,
hepatitis B virus X protein, which is involved in the initia-
tion and progression of HCC downregulates miR-15a/16
expression, suggesting that reintroduction of these miRNAs
may be an effective treatment of hepatitis-B-virus-related
chronic liver diseases.81 MiR-15a and miR-16-1 act tumor
suppressive, at least in part, by promoting apoptosis and
cell cycle inhibition via targeting the anti-apoptotic pro-
tein Bcl282 and cell cycle regulators, including CDK6 and
cyclin D1, respectively.83,84 Recently, we demonstrated that
miR- 15a/16-1 also inhibit EMT, invasion, and metastasis
of CRC cells by directly targeting the EMT-TF AP4.85
Interestingly, AP4 itself is a repressor of the DLEU2 gene.
We showed that miR-15a/16-1 and the EMT-inducing factor
AP486 form a double-negative feedback loop that stabilizes
low expression of miR-15a/16-1 and elevated expression of
AP4 in invasive CRC cells and tumors, thereby ultimately
promoting CRC metastasis (Figure 3C).85
miR-145
p53 controls the expression of miR-145 by two mecha-
nisms: first, p53 directly induces the transcription of
the miR-145 gene,35 and second, p53 enhances miR-145
maturation via modulation of Drosha-mediated miRNA
processing.28,87 In line with a regulation by p53, expres-
sion of miR-145 is significantly lower in various tumors
that harbor p53 mutations, including esophageal, gastric,
pancreatic, colorectal, and bladder cancers.88–92 Accord-
ingly, ectopic miR-145 suppresses migration, invasion,
and metastasis of gastric and colorectal cancer cells.89,93
Moreover, therapeutic polyethylenimine-mediated rein-
troduction of miR-145 reduces proliferation and increases
apoptosis of CRC cells in xenograft mouse models.94 The
tumor suppressing properties of miR-145 can be partially
attributed to the repression of MYC, which represents a
direct target of miR-145.35 Similar to miR-34, miR-200, and
miR-15a/16-1, miR-145 has also been shown to represent a
mediator of p53-induced MET, the reversion of EMT (Fig-
ure 3C).95 Another important oncogenic target of miR-145
is KRAS.96 Interestingly, activated KRAS also represses
miR-145 via RREB1, thereby forming a feed-forward
loop that potentiates RAS signaling. Accordingly, loss
of miR-145 is frequently observed in KRAS mutant pan-
creatic cancers, and restoration of these miRNAs inhibits
tumorigenesis.96 Xu et al showed that miR-145 negatively
regulates the pluripotency factors OCT4, SOX2, and KLF4,
and thereby represses self-renewal and induces differentia-
tion of human embryonic stem cells.97 Moreover, the same
group reported that the miR-145 promoter is bound and
repressed by OCT4, thereby forming a negative feedback
loop.97 Loss of p53 leads to increased generation of induced
pluripotent stem cells and expansion of cancer stem cells.98
This effect might at least in part be due to the lack of p53-
induced miR-145 expression and consequent upregulation
of OCT4. Finally, like miR-34a targets MDM4, miR-145
directly targets and represses the p53 inhibitor MDM2.99
The result is another positive feed-forward loop that leads
to stabilization of p53 and elevated expression of p53-
induced miRNAs (Figure 3A).
The miR-192/miR-194/
miR-215 family
MiR-192, miR-194, and miR-215 are encoded by two clusters
located at two different sites: The miR-194-1/miR-215 cluster
Clinical and Experimental Gastroenterology 2014:7
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
404
Rokavec et al
on chromosome 1 (1q41) and the miR-192/miR-194-2 clus-
ter on chromosome 11 (11q13.1). Both clusters are directly
induced by p53.34,100 Interestingly, miR-194-1 and miR-194-2
have the same mature sequence, although they are derived
from two different precursors on two chromosomal locations.
MiR-192 and miR-215 have the same seed sequence, whereas
the seed sequence of miR-194 differs. All three miRNAs dis-
play decreased expression in colorectal tumors.34 Furthermore,
low expression of miR-194 and miR-215 significantly corre-
lates with a high probability of relapse and shorter survival in
colorectal patients.101 MiR-192, miR-194, and miR-215 regu-
late cell cycle progression and proliferation via the repression
of functionally important targets, such as CDC7, MAD2L1,
and CUL5.102 Moreover, miR-192 suppresses liver metasta-
sis of CRC cells by targeting BCL2, ZEB2, and VEGF-A,103
while miR-194 inhibits EMT in endometrial cells by targeting
BMI-1.104 Importantly, similar to other p53-induced miRNAs,
miR-192, miR-194, and miR-215 also directly target MDM2
and therefore interfere with the autoregulatory MDM2/p53
loop (Figure 3A).100
The miR-200 family
The two genes that give rise to the miR-200c/141 and the
200a/200b/429 miRNAs represent direct p53 target genes.32
The members of the miR-200 family are tumor suppress-
ing miRNAs and several studies showed that they play a
crucial role in regulating the balance between EMT and
MET by forming a double-negative feedback loop with
their targets, the EMT-inducers ZEB1 and ZEB2 (Figure
3C).52,105 Moreover, miR-200c also suppresses stemness
by targeting the stem cell factors KLF4, SOX2, and the
polycomb repressor BMI-1.106,107 Several studies reported
that elevated levels of cell-free, circulating miR-200c and
miR-200a in the blood of colorectal, gastric, and esophageal
cancer patients are associated with increased tumor stage,
presence of metastases, and poor survival.108–112 At first
sight, this data seems contradictory, since functional studies
showed that miR-200s repress EMT, invasion, and metas-
tasis. Yet, recent studies showed that during the formation
of metastases, cancer cells undergo MET and re-express
EMT-suppressing genes and miRNAs.113 Therefore, elevated
levels of EMT-suppressing circulating miRNAs, such as
miR-200c, might originate from metastases and indicate
metastatic dissemination.
miR-107
The miRNA miR-107 is encoded by an intron of the p53-
induced pantothenate kinase 1 (PANK1) gene.36,114 Several
studies showed that ectopic expression of miR-107 enhances
EMT, migration, and promotes metastatic dissemina-
tion, whereas the loss of miR-107 represses migration
and metastasis of colorectal, breast, and gastric cancer
cells.115–118 In line with these observations, expression of
miR-107 is higher in gastric tumors compared with adjacent
normal tissue.119 Moreover, high expression of miR-107
correlates with lymph node and distant metastasis as well
as poor survival of colorectal, breast, and gastric cancer
patients.115–120 The pro-metastatic properties of miR-107
are mediated by repression of its targets, the metastasis
suppressors DAPK and KLF4.117 Furthermore, Martello
et al showed that miR-107 targets and represses DICER1,
a key component of the miRNA processing machinery,
thereby attenuating global miRNA production.115 Therefore,
elevated levels of miR-107 in tumors may contribute to the
global reduction of miRNA abundance that was observed
in various cancer types.121 In addition to regulation of
mRNA targets, miR-107 can also directly interact with and
negatively regulate the let-7 miRNA.116 Accordingly, miR-
107 increased the tumorigenic and metastatic potential of
human breast cancer cell lines in xenograft mouse models
via inhibition of let-7 and upregulation of let-7 targets.116
However, others have shown that miR-107 also has tumor
suppressing functions by inhibiting cell proliferation and
migration of breast cancer, gastric cancer, and glioma
cells.120,122,123 These tumor suppressing effects could be par-
tially attributed to the miR-107-mediated repression of the
response to hypoxia and angiogenesis via targeting of HIF
1β, resulting in a decreased supply of oxygen and nutrients
and subsequent inhibition of tumor growth.114 The decrease
in functional HIF1α–HIF1β dimers after p53-mediated acti-
vation of miR-107 may suppress glycolysis under hypoxic
conditions. These results indicate that p53-deficient tumors
may be resistant to hypoxia not only because of decreased
apoptosis and senescence, but also because of increased
HIF1 signalling due to the decrease in miR-107 which
results in metabolic and angiogenic adaptation. Altogether,
the majority of the current data suggests that miR-107 is
an oncogenic miRNA that promotes EMT, migration, and
metastasis. However, these observations are at first sight not
compatible with the induction of miR-107 by p53, which
would be expected to mediate tumor suppressive functions.
A possible explanation may be that p53 induces miR-107
and thereby downregulates DICER1 to limit the production
of p53-induced miRNAs, which would otherwise lead to
an unrestrained induction of p53 because of the positive
feedback loops these often form with p53.
Clinical and Experimental Gastroenterology 2014:7 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
405
The p53/microRNA axis in GI cancer
p53-repressed miRNAs
p53 also directly represses certain miRNAs, including
miR-224,124 miR-502,125 and the miR-17-92 cluster.126
However, this type of regulation seems to occur less fre-
quently than the induction of tumor suppressive miRNAs
by p53. As expected, miRNAs repressed by p53 mostly
have oncogenic functions. The miR-17-92 primary
transcript encodes the miRNAs miR-17, miR-18a, miR-
19a, miR-20a, miR-19b-1, and miR-92a-1. The miR-17-92
clusters undergo genomic amplification and display elevated
expression in various cancer entities, including colon can-
cer.127 The miRNAs of the miR-17-92 family promote cell
proliferation, increase angiogenesis, promote cell survival,
and exhibit strong protumorigenic activities in multiple
mouse tumor models.128 Yan et al showed that miR-17-92
is repressed upon hypoxia via direct interaction of p53 with
the miR-17-92 promoter.126 Due to the strong cell survival
promoting properties of miR-17-92 family members, it is
likely that the repression of miR-17-92 expression by p53
plays a role in p53-induced apoptosis.
Regulation of p53 by miRNAs
p53 not only regulates the expression and processing of
miRNAs, but is also under the control of certain miRNAs.
Several miRNAs repress the translation of TP53 mRNA
by directly binding to its 3-UTR (Tables 1 and 3). Since
these miRNAs diminish the tumor suppressive activity
of p53, they often represent oncomirs. Accordingly, they
often exhibit elevated expression in tumors. Similar to the
p53-regulated miRNAs the expression of p53-regulating
miRNAs is frequently altered in GI tumors (Tables 1 and 3).
The first miRNA that was characterized as a direct suppres-
sor of p53 was miR-125b. Le et al. Showed that miR-125b
is a negative regulator of p53 expression and p53-induced
apoptosis during development and stress response.129
Furthermore, it has been shown that miR-125 targets several
Table 3 Compilation of miRNAs that directly target p53 and their alterations in GI cancers
miRNA Validated by Clinical and pathological associations in GI cancers
miR-25 Luc reporter (mut), qPCR, WB134 CRC: upregulated in tumors; Upregulation associated with invasion, metastasis, poor OS146
EC: upregulated in tumors and serum;233 Upregulation associated with pN and tumor stage137,145
GC: upregulated in serum234 and tumors;235 Upregulation associated with tumor progression236,237
miR-30d Luc reporter (mut), qPCR, WB134 CRC, PaC: gene amplication148
HCC: upregulated in tumors; Upregulation associated with metastasis238
miR-33 Luc reporter (mut), WB135 NA
miR-98 Luc reporter (mut), WB143 EC: downregulated in tumors; Downregulation associated with tumor stage and pN239
miR-125a Luc reporter (mut), WB142 EC: downregulation associated with tumor progression240
GC: downregulation associated with invasion, metastasis, poor OS240–242
HCC: downregulated in tumors; Downregulation associated with tumor stage and metastasis243,244
miR-125b Luc reporter (mut), WB129 CRC: upregulation associated with tumor size, invasion, poor OS241
EC: downregulation associated with tumor progression240
GC: upregulation associated with tumor progression;236 Downregulation associated with tumor
progression240
HCC: upregulated in serum;131 Downregulated in tumors244,245
PaC: upregulated in tumors246
miR-150 Luc reporter (mut), WB138 CRC: downregulated in tumors; Downregulation associated with poor OS247
EC: downregulated in tumors; Downregulation associated with invasion, metastasis, and poor OS248
GC: upregulated in tumors249
PaC: downregulated in tumors250
miR-214 Luc reporter (mut), WB141 CRC: upregulation associated with poor OS236
EC: upregulated in tumors;251 Downregulated in tumors239
GC: upregulated in tumors; Upregulation associated with poor OS252
HCC: downregulated in tumors; Downregulation associated with poor RFS and OS253–255
PaC: upregulated in tumors78
miR-375 Luc reporter (mut), WB139 EC: downregulated in tumors and serum; Downregulation associated with poor OS145
PaC: downregulated in tumors236
miR-380 Luc reporter, WB136 NA
miR-504 Luc reporter (mut), WB133 NA
miR-1285 Luc reporter (mut), qPCR, WB140 NA
Abbreviations: CRC, colorectal cancer; EC, esophageal cancer; GC, gastric cancer; GI, gastrointestinal; HCC, hepatocellular cancer; Luc reporter, Luciferase reporter
assay with p53 3-UTR; miRNA, microRNA; mut, mutation in the miRNA seed sequence; NA, not applicable or not analyzed; OS, overall survival; PaC, pancreatic cancer;
pN, nodal status; qPCR, quantitative real-time polymerase chain reaction; RFS, relapse free survival; WB, Western blot.
Clinical and Experimental Gastroenterology 2014:7
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
406
Rokavec et al
additional components of the p53 network, which include
both regulators of apoptosis like Bak1, Igfbp3, Itch, Puma,
Prkra, Tp53inp1, and Zac1, and components of the cell
cycle machinery such as cyclin C, Cdc25c, Cdkn2c, Edn1,
Ppp1ca, and Sel1l.130 The authors proposed that by regulat-
ing proliferative and apoptotic genes, miR-125b buffers
and fine-tunes the activity of the p53 network in order to
control the balance between proliferation and apoptosis.
Recently, it was shown that the levels of miR-125b are sig-
nificantly higher in the serum of patients with hepatitis-B-
virus-positive HCC and therefore circulating miR-125b may
represent a potential non-invasive marker for HCC.131 More-
over, elevated expression of miR-125 was also associated
with increased tumor size, enhanced invasion, and poor prog-
nosis in CRC patients.132 Another miRNA that negatively
regulates p53 expression via two seed-matching sequences
in the human TP53 3-UTR is miR-504.133 Accordingly, ecto-
pic expression of miR-504 reduced p53 protein levels and
impaired p53 functions, especially p53-mediated apoptosis
and G1-arrest in response to stress. Furthermore, ectopic
expression of miR-504 promoted tumorigenicity of colon
cancer cells in mice.133 Additionally, miR-25, miR-30d,
miR-33, miR-98, miR-150, miR-214, miR-375, miR-380,
and miR-1285 also downregulate p53 protein levels through
seed-matching sequences in the 3-UTR of TP53.134–144
Accordingly, ectopic expression of these miRNAs suppresses
p53 expression and induces phenotypes that are consistent
with a decrease in p53 function, such as reduced apoptosis
and senescence, and increased invasion and stem cell self-
renewal.134–137 miR-25 levels were increased in esophageal
tumors and serum of esophageal cancer patients displayed
elevated levels of circulating miR-25.145 Moreover, expres-
sion of miR-25 was significantly higher in colorectal tumors
and elevated levels of miR-25 were associated with increased
tumor invasion, lymph node metastasis, distant metastasis,
TNM (tumor, node, metastasis status based classification)
stage, and poor survival of CRC patients.146 MiR-30d is
an important regulator of autophagy,147 and interestingly,
amplification of the MIR30D gene was found in 30% of
1,283 analyzed solid tumors, including bladder, colorectal,
and pancreatic cancer.148
In addition to the direct repression of p53 by miRNAs,
several miRNAs also regulate the expression of p53
indirectly. As described above, the expression of the p53
inhibitors MDM2 and MDM4 is directly repressed by sev-
eral p53-induced miRNAs: MDM2 is a target of miR-145,
miR-192/194/215, miR-605, and miR-29b, whereas MDM4
is targeted by the miR-34 family members (Figure 3A).
Therefore, p53-mediated induction of these miRNAs results
in a positive feedback and enhanced p53 activation. In addi-
tion, miRNAs of the miR-29 family target the expression of
other negative regulators of p53, such as Cdc42, PPM1D,
and the regulatory subunit of phosphatidylinositol-3 kinase
(PI3K), p85α, and thereby indirectly enhance the levels and
activity of p53.149,150 In addition, miR-29 is also directly
induced by p53,149 thereby forming a positive feedback
loop that is activated during aging and DNA damage,
and reinforces p53 effector functions, such as apoptosis
and senescence. The members of the miR-29 family are
aberrantly expressed in various tumors, including gastric
cancer and HCC.151–153 Moreover, low expression of miR-29
in HCC was associated with decreased survival.154 Fur-
thermore, miR-122, which is expressed exclusively in the
liver, is frequently downregulated in liver cancer.155 It was
shown that miR-122 stabilizes and therefore increases p53
protein levels and activity via downregulation of its target
cyclin G1.156 Repression of cyclin G1 results in decreased
recruitment of the PP2A phosphatase to the p53-inhibitor
MDM2. The resulting decrease in MDM2 activity leads to
activation of p53.157 Interestingly, Cyclin G1 is also directly
induced by p53 and therefore forms a negative feedback
loop with p53.158 However, therapeutic treatment with
miR-122 mimtics might abrogate this loop by downregula-
tion of cyclin G1. Indeed, it has been shown that ectopic
miR-122 expression increases the sensitivity of HCC cell
lines to the chemotherapeutic agent doxorubicin, which is
known to induce p53.156 However, it should be noted that
miR-122 increases chemosensitivity also in the absence of
wild-type p53 and therefore seems to have p53-independent
functions.
Conclusion and outlook
Although numerous links between p53 and miRNAs have
been identified, we have only begun to understand the
complex interplay between p53 and miRNAs in tumor
suppression. Therefore, additional efforts are necessary to
uncover more details of the p53/miRNA network. Since p53
has many functions in tumor suppression, future research
should focus on identifying which miRNA is responsible
for mediating specific p53 functions. It would also be of
interest to investigate whether the complete spectrum of
tumor suppressing functions of p53, which is frequently
lost in tumors, can be restored by the introduction of spe-
cific combinations of p53-induced miRNAs. So far, the
majority of studies have rather focused on the identifica-
tion and characterization of single p53-regulated miRNAs.
Clinical and Experimental Gastroenterology 2014:7 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
407
The p53/microRNA axis in GI cancer
A feasible strategy for a comprehensive, genome-wide
identification of p53-regulated miRNAs and their target
genes has been recently described by us.159 This strategy
employs a combination of various unbiased genome-wide
next generation sequencing screens to simultaneously
identify and characterize p53-regulated miRNAs and their
targets. By now, knockout mouse models of single p53-
induced miRNAs have not fully recapitulated the cancerous
phenotype of p53 knockout mice, which is characterized
by the early onset of lymphomas.160 Therefore, it would
be interesting to generate genetically modified mice
that simultaneously lack multiple p53-related miRNAs
to investigate whether loss of certain combinations of
miRNAs fully or at least partially mimics the phenotype
of p53-knockout mice. Complementarily, p53 knockout
mice could be treated with a cocktail of p53-induced
miRNAs to investigate whether certain combinations of
miRNAs can suppress the tumor promoting effects of p53
loss. In light of the central role of p53-regulated miRNAs
or p53-regulating miRNAs for tumor suppression, the
introduction of these miRNAs or of miRNA antagonists
into tumor cells represents an exciting possibility for novel
cancer-therapeutic approaches. Such therapies could also
be performed in combination with standard anticancer
therapies as has been already shown for miR-34a, which
sensitized gastric cancer cells to the chemotherapeutic
drugs docetaxel, gemcitabine, cisplatin, and doxorubicin.63
A major obstacle for such approaches is the currently low
efficiency of miRNA delivery into tumor cells. Therefore,
further research is needed to develop more efficient strate-
gies for in vivo miRNA delivery. Promisingly, improved
delivery using novel nanoparticles was recently employed
to show that a combination of miR-34a mimics and siRNAs
directed at mutant oncogenes is more effective than either
RNA alone in a pre-clinical mouse model of lung cancer.161
Since p53-regulated miRNAs are often inactivated by CpG
methylation in tumors, establishment of routine protocols
for detection of methylation of selected promoters of
miRNA-encoding genes in body fluids may represent an
important aspect of future GI cancer diagnostics.
Disclosure
The authors report no conflicts of interest in this work.
References
1. International Agency for Research on Cancer. GLOBOCAN 2012:
Estimated Cancer Incidence, Mortality and Prevalence Worldwide in 2012.
Geneva: World Health Organization; 2014. Available from: http://globocan.
iarc.fr/Pages/fact_sheets_population.aspx. Accessed July 30, 2014.
2. International Agency for Research on Cancer. IARC TP53 Databse.
Geneva: World Health Organization; 2014. Available from: http://p53.
iarc.fr/TP53SomaticMutations.aspx. Accessed July 30, 2014.
3. Vogelstein B, Lane D, Levine AJ. Surfing the p53 network. Nature.
2000;408(6810):307–310.
4. Oren M. Decision making by p53: life, death and cancer. Cell Death
Differ. 2003;10(4):431–442.
5. Goh AM, Coffill CR, Lane DP. The role of mutant p53 in human cancer.
J Pathol. 2011;223(2):116–126.
6. Lane D, Levine A. p53 Research: the past thirty years and the next
thirty years. Cold Spring Harb Perspect Biol. 2010;2(12):a000893.
7. el-Deiry WS, Kern SE, Pietenpol JA, Kinzler KW, Vogelstein B.
Definition of a consensus binding site for p53. Nat Genet. 1992;1(1):
45–49.
8. Hermeking H. p53 enters the microRNA world. Cancer Cell. 2007;
12(5):414–418.
9. Hermeking H. MicroRNAs in the p53 network: micromanagement of
tumour suppression. Nat Rev Cancer. 2012;12(9):613–626.
10. Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function.
Cell. 2004;116(2):281–297.
11. Krol J, Loedige I, Filipowicz W. The widespread regulation of microRNA
biogenesis, function and decay. Nat Rev Genet. 2010;11(9):597–610.
12. Yates LA, Norbury CJ, Gilbert RJ. The long and short of microRNA.
Cell. 2013;153(3):516–519.
13. Friedman RC, Farh KK, Burge CB, Bartel DP. Most mammalian
mRNAs are conserved targets of microRNAs. Genome Res. 2009;19(1):
92–105.
14. Fassan M, Croce CM, Rugge M. miRNAs in precancerous lesions of the
gastrointestinal tract. World J Gastroenterol. 2011;17(48): 5231–5239.
15. Song B, Ju J. Impact of miRNAs in gastrointestinal cancer diagnosis
and prognosis. Expert Rev Mol Med. 2010;12:e33.
16. Boni V, Bandres E, Zarate R, Colucci G, Maiello E, Garcia-Foncillas J.
MicroRNAs as a new potential therapeutic opportunity in gastrointes-
tinal cancer. Oncology. 2009;77 Suppl 1:75–89.
17. Vicentini C, Fassan M, D’Angelo E, et al. Clinical application of
microRNA testing in neuroendocrine tumors of the gastrointestinal
tract. Molecules. 2014;19(2):2458–2468.
18. Macha MA, Seshacharyulu P, Krishn SR, et al. MicroRNAs (miRNA)
as Biomarker(s) for Prognosis and Diagnosis of Gastrointestinal (GI)
Cancers. Curr Pharm Des. Epub January 28, 2014.
19. Ajit SK. Circulating microRNAs as biomarkers, therapeutic targets,
and signaling molecules. Sensors (Basel). 2012;12(3):3359–3369.
20. Blanco-Calvo M, Calvo L, Figueroa A, Haz-Conde M, Antón-Aparicio L,
Valladares-Ayerbes M. Circulating microRNAs: molecular microsensors
in gastrointestinal cancer. Sensors (Basel). 2012;12(7):9349–9362.
21. Bommer GT, Gerin I, Feng Y, et al. p53-mediated activation of
miRNA34 candidate tumor-suppressor genes. Curr Biol. 2007;17(15):
1298–1307.
22. Chang TC, Wentzel EA, Kent OA, et al. Transactivation of miR-34a by
p53 broadly influences gene expression and promotes apoptosis. Mol
Cell. 2007;26(5):745–752.
23. Corney DC, Flesken-Nikitin A, Godwin AK, Wang W, Nikitin AY.
MicroRNA-34b and MicroRNA-34c are targets of p53 and cooperate in
control of cell proliferation and adhesion-independent growth. Cancer
Res. 2007;67(18):8433–8438.
24. He L, He X, Lim LP, et al. A microRNA component of the p53 tumour
suppressor network. Nature. 2007;447(7148):1130–1134.
25. Raver-Shapira N, Marciano E, Meiri E, et al. Transcriptional activa-
tion of miR-34a contributes to p53-mediated apoptosis. Mol Cell.
2007;26(5):731–743.
26. Tarasov V, Jung P, Verdoodt B, et al. Differential regulation of microRNAs by
p53 revealed by massively parallel sequencing: miR-34a is a p53 target that
induces apoptosis and G1-arrest. Cell Cycle. 2007;6(13):1586–1593.
27. Tazawa H, Tsuchiya N, Izumiya M, Nakagama H. Tumor-suppressive
miR-34a induces senescence-like growth arrest through modulation
of the E2F pathway in human colon cancer cells. Proc Natl Acad Sci
U S A. 2007;104(39):15472–15477.
Clinical and Experimental Gastroenterology 2014:7
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
408
Rokavec et al
28. Suzuki HI, Yamagata K, Sugimoto K, Iwamoto T, Kato S, Miyazono K.
Modulation of microRNA processing by p53. Nature. 2009;460(7254):
529–533.
29. Mudhasani R, Zhu Z, Hutvagner G, et al. Loss of miRNA biogenesis
induces p19Arf-p53 signaling and senescence in primary cells. J Cell
Biol. 2008;181(7):1055–1063.
30. Su X, Chakravarti D, Cho MS, et al. TAp63 suppresses metasta-
sis through coordinate regulation of Dicer and miRNAs. Nature.
2010;467(7318):986–990.
31. Léveillé N, Elkon R, Davalos V, et al. Selective inhibition of microRNA
accessibility by RBM38 is required for p53 activity. Nat Commun.
2011;2:513.
32. Kim T, Veronese A, Pichiorri F, et al. p53 regulates epithelial-
mesenchymal transition through microRNAs targeting ZEB1 and ZEB2.
J Exp Med. 2011;208(5):875–883.
33. Fabbri M, Bottoni A, Shimizu M, et al. Association of a microRNA/
TP53 feedback circuitry with pathogenesis and outcome of B-cell
chronic lymphocytic leukemia. JAMA. 2011;305(1):59–67.
34. Braun CJ, Zhang X, Savelyeva I, et al. p53-Responsive micrornas
192 and 215 are capable of inducing cell cycle arrest. Cancer Res.
2008;68(24):10094–10104.
35. Sachdeva M, Zhu S, Wu F, et al. p53 represses c-Myc through induction
of the tumor suppressor miR-145. Proc Natl Acad Sci U S A. 2009;
106(9):3207–3212.
36. Böhlig L, Friedrich M, Engeland K. p53 activates the PANK1/miRNA-
107 gene leading to downregulation of CDK6 and p130 cell cycle
proteins. Nucleic Acids Res. 2011;39(2):440–453.
37. Wang J, He Q, Han C, et al. p53-facilitated miR-199a-3p regulates
somatic cell reprogramming. Stem Cells. 2012;30(7):1405–1413.
38. Roy S, Levi E, Majumdar AP, Sarkar FH. Expression of miR-34 is lost
in colon cancer which can be re-expressed by a novel agent CDF. J
Hematol Oncol. 2012;5:58.
39. Cui X, Zhao Z, Liu D, et al. Inactivation of miR-34a by aberrant CpG
methylation in Kazakh patients with esophageal carcinoma. J Exp Clin
Cancer Res. 2014;33:20.
40. Stánitz E, Juhász K, Tóth C, Gombos K, Natali PG, Ember I. Evaluation
of MicroRNA expression pattern of gastric adenocarcinoma associated
with socioeconomic, environmental and lifestyle factors in northwestern
Hungary. Anticancer Res. 2013;33(8):3195–3200.
41. Xie K, Liu J, Chen J, et al. Methylation-associated silencing of microRNA-
34b in hepatocellular carcinoma cancer. Gene. 2014;543(1):101–107.
42. Wang R, Ma J, Wu Q, et al. Functional role of miR-34 family in human
cancer. Curr Drug Targets. 2013;14(10):1185–1191.
43. Rokavec M, Li H, Jiang L, Hermeking H. The p53/miR-34 axis in
development and disease. J Mol Cell Biol. 2014;6(3):214–230.
44. Mandke P, Wyatt N, Fraser J, Bates B, Berberich SJ, Markey MP.
MicroRNA-34a modulates MDM4 expression via a target site in the
open reading frame. PLoS One. 2012;7(8):e42034.
45. Okada N, Lin CP, Ribeiro MC, et al. A positive feedback between
p53 and miR-34 miRNAs mediates tumor suppression. Genes Dev.
2014;28(5):438–450.
46. Yamakuchi M, Ferlito M, Lowenstein CJ. miR-34a repression of SIRT1 reg-
ulates apoptosis. Proc Natl Acad Sci U S A. 2008;105(36): 13421–13426.
47. Choi SE, Fu T, Seok S, et al. Elevated microRNA-34a in obesity reduces
NAD+ levels and SIRT1 activity by directly targeting NAMPT. Aging
Cell. 2013;12(6):1062–1072.
48. Menssen A, Hydbring P, Kapelle K, et al. The c-MYC oncoprotein,
the NAMPT enzyme, the SIRT1-inhibitor DBC1, and the SIRT1
deacetylase form a positive feedback loop. Proc Natl Acad Sci U S A.
2012;109(4):E187–E196.
49. Marshall GM, Liu PY, Gherardi S, et al. SIRT1 promotes N-Myc
oncogenesis through a positive feedback loop involving the effects of
MKP3 and ERK on N-Myc protein stability. PLoS Genet. 2011;7(6):
e1002135.
50. Siemens H, Jackstadt R, Hünten S, et al. miR-34 and SNAIL form
a double-negative feedback loop to regulate epithelial-mesenchymal
transitions. Cell Cycle. 2011;10(24):4256–4271.
51. Burk U, Schubert J, Wellner U, et al. A reciprocal repression between
ZEB1 and members of the miR-200 family promotes EMT and invasion
in cancer cells. EMBO Rep. 2008;9(6):582–589.
52. Gregory PA, Bert AG, Paterson EL, et al. The miR-200 family and
miR-205 regulate epithelial to mesenchymal transition by targeting
ZEB1 and SIP1. Nat Cell Biol. 2008;10(5):593–601.
53. Ahn YH, Gibbons DL, Chakravarti D, et al. ZEB1 drives prometastatic
actin cytoskeletal remodeling by downregulating miR-34a expression.
J Clin Invest. 2012;122(9):3170–3183.
54. Hahn S, Jackstadt R, Siemens H, Hünten S, Hermeking H. SNAIL
and miR-34a feed-forward regulation of ZNF281/ZBP99 pro-
motes epithelial-mesenchymal transition. EMBO J. 2013;32(23):
3079–3095.
55. Rokavec M, Öner MG, Li H, et al. IL-6R/STAT3/miR-34a feedback
loop promotes EMT-mediated colorectal cancer invasion and metastasis.
J Clin Invest. 2014;124(4):1853–1867.
56. Bader AG. miR-34 a microRNA replacement therapy is headed to the
clinic. Front Genet. 2012;3:120.
57. Pramanik D, Campbell NR, Karikari C, et al. Restitution of tumor
suppressor microRNAs using a systemic nanovector inhibits pancreatic
cancer growth in mice. Mol Cancer Ther. 2011;10(8):1470–1480.
58. Craig VJ, Tzankov A, Flori M, Schmid CA, Bader AG, Müller A.
Systemic microRNA-34a delivery induces apoptosis and abrogates
growth of diffuse large B-cell lymphoma in vivo. Leukemia. 2012;
26(11):2421–2424.
59. Wiggins JF, Ruffino L, Kelnar K, et al. Development of a lung cancer
therapeutic based on the tumor suppressor microRNA-34. Cancer Res.
2010;70(14):5923–5930.
60. Mirna Therapeutics, Inc. [homepage on the Internet]. Austin: Mirna
Therapeutics, Inc.; 2014. Available from: http://www.mirnarx.com/.
Accessed April 30, 2014.
61. Fujita Y, Kojima K, Hamada N, et al. Effects of miR-34a on cell growth
and chemoresistance in prostate cancer PC3 cells. Biochem Biophys Res
Commun. 2008;377(1):114–119.
62. Vin all RL, Rip oll A Z, Wang S, Pan C-X, deVere Whit e RW.
MiR-34a chemosensitizes bladder cancer cells to cisplatin
treatment regardless of p53-Rb pathway status. Int J Cancer.
2012;130(11):2526–2538.
63. Ji Q, Hao X, Meng Y, et al. Restoration of tumor suppressor miR-34
inhibits human p53-mutant gastric cancer tumorspheres. BMC Cancer.
2008;8:266.
64. Ji Q, Hao X, Zhang M, et al. MicroRNA miR-34 inhibits human pan-
creatic cancer tumor-initiating cells. PLoS One. 2009;4(8):e6816.
65. Siemens H, Jackstadt R, Kaller M, Hermeking H. Repression of
c-Kit by p53 is mediated by miR-34 and is associated with reduced
chemoresistance, migration and stemness. Oncotarget. 2013;4(9):
1399–1415.
66. Lodygin D, Tarasov V, Epanchintsev A, et al. Inactivation of miR-34a
by aberrant CpG methylation in multiple types of cancer. Cell Cycle.
2008;7(16):2591–2600.
67. Vogt M, Munding J, Grüner M, et al. Frequent concomitant inactivation
of miR-34a and miR-34b/c by CpG methylation in colorectal, pancre-
atic, mammary, ovarian, urothelial, and renal cell carcinomas and soft
tissue sarcomas. Virchows Arch. 2011;458(3):313–322.
68. Toyota M, Suzuki H, Sasaki Y, et al. Epigenetic silencing of
microRNA-34b/c and B-cell translocation gene 4 is associated with
CpG island methylation in colorectal cancer. Cancer Res. 2008;68(11):
4123–4132.
69. Suzuki H, Yamamoto E, Nojima M, et al. Methylation-associated
silencing of microRNA-34b/c in gastric cancer and its involvement in
an epigenetic field defect. Carcinogenesis. 2010;31(12):2066–2073.
70. Siemens H, Neumann J, Jackstadt R, et al. Detection of miR-34a pro-
moter methylation in combination with elevated expression of c-Met
and β-catenin predicts distant metastasis of colon cancer. Clin Cancer
Res. 2013;19(3):710–720.
71. Hermeking H. The miR-34 family in cancer and apoptosis. Cell Death
Differ. 2010;17(2):193-199.
Clinical and Experimental Gastroenterology 2014:7 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
409
The p53/microRNA axis in GI cancer
72. Kong D, Heath E, Chen W, et al. Epigenetic silencing of miR-34a in
human prostate cancer cells and tumor tissue specimens can be reversed
by BR-DIM treatment. Am J Transl Res. 2012;4(1):14–23.
73. Östling P, Leivonen SK, Aakula A, et al. Systematic analysis of
microRNAs targeting the androgen receptor in prostate cancer cells.
Cancer Res. 2011;71(5):1956–1967.
74. Wu XD, Song YC, Cao PL, et al. Detection of miR-34a and miR- 34b/c
in stool sample as potential screening biomarkers for noninvasive
diagnosis of colorectal cancer. Med Oncol. 2014;31(4):894.
75. Calin GA, Dumitru CD, Shimizu M, et al. Frequent deletions and down-
regulation of micro- RNA genes miR15 and miR16 at 13q14 in chronic lym-
phocytic leukemia. Proc Natl Acad Sci U S A. 2002;99(24): 15524–15529.
76. Klein U, Lia M, Crespo M, et al. The DLEU2/miR-15a/16-1 cluster
controls B cell proliferation and its deletion leads to chronic lymphocytic
leukemia. Cancer Cell. 2010;17(1):28–40.
77. Qian J, Jiang B, Li M, Chen J, Fang M. Prognostic significance of
microRNA-16 expression in human colorectal cancer. World J Surg.
2013;37(12):2944–2949.
78. Zhang XJ, Ye H, Zeng CW, He B, Zhang H, Chen YQ. Dysregulation of
miR-15a and miR-214 in human pancreatic cancer. J Hematol Oncol.
2010;3:46.
79. Ma Q, Wang X, Li Z, et al. microRNA-16 represses colorectal cancer
cell growth in vitro by regulating the p53/survivin signaling pathway.
Oncol Rep. 2013;29(4):1652–1658.
80. Dai L, Wang W, Zhang S, et al. Vector-based miR-15a/16-1 plasmid
inhibits colon cancer growth in vivo. Cell Biol Int. 2012;36(8):
765–770.
81. Wu G, Yu F, Xiao Z, et al. Hepatitis B virus X protein downregulates
expression of the miR-16 family in malignant hepatocytes in vitro.
Br J Cancer. 2011;105(1):146–153.
82. Cimmino A, Calin GA, Fabbri M, et al. miR-15 and miR-16 induce
apoptosis by targeting BCL2. Proc Natl Acad Sci U S A. 2005;102(39):
13944–13949.
83. Liu Q, Fu H, Sun F, et al. miR-16 family induces cell cycle arrest by
regulating multiple cell cycle genes. Nucleic Acids Res. 2008;36(16):
5391–5404.
84. Bonci D, Coppola V, Musumeci M, et al. The miR-15a-miR-16-1 cluster
controls prostate cancer by targeting multiple oncogenic activities. Nat
Med. 2008;14(11):1271–1277.
85. Shi L, Jackstadt R, Siemens H, Li H, Kirchner T, Hermeking H. p53-
induced miR-15a/16-1 and AP4 form a double-negative feedback loop to
regulate epithelial-mesenchymal transition and metastasis in colorectal
cancer. Cancer Res. 2014;74(2):532–542.
86. Jackstadt R, Röh S, Neumann J, et al. AP4 is a mediator of epithelial-
mesenchymal transition and metastasis in colorectal cancer. J Exp Med.
2013;210(7):1331–1350.
87. Suzuki K, Matsubara H. Recent advances in p53 research and cancer
treatment. J Biomed Biotechnol. 2011;2011:978312.
88. Kano M, Seki N, Kikkawa N, et al. miR-145, miR-133a and miR-133b:
Tumor-suppressive miRNAs target FSCN1 in esophageal squamous cell
carcinoma. Int J Cancer. 2010;127(12):2804–2814.
89. Gao P, Xing a-Y, Zhou GY, et al. The molecular mechanism of
microRNA-145 to suppress invasion-metastasis cascade in gastric
cancer. Oncogene. 2013;32(4):491–501.
90. Papaconstantinou IG, Manta A, Gazouli M, et al. Expression of microR-
NAs in patients with pancreatic cancer and its prognostic significance.
Pancreas. 2013;42(1):67–71.
91. Slaby O, Svoboda M, Fabian P, et al. Altered expression of miR-21,
miR-31, miR-143 and miR-145 is related to clinicopathologic features
of colorectal cancer. Oncology. 2007;72(5–6):397–402.
92. Ratert N, Meyer HA, Jung M, et al. miRNA profiling identifies candidate
mirnas for bladder cancer diagnosis and clinical outcome. J Mol Diagn.
2013;15(5):695–705.
93. Pagliuca a, Valvo C, Fabrizi E, et al. Analysis of the combined action
of miR-143 and miR-145 on oncogenic pathways in colorectal cancer
cells reveals a coordinate program of gene repression. Oncogene.
2013;32(40):4806–4813.
94. Ibrahim AF, Weirauch U, Thomas M, Grünweller A, Hartmann RK,
Aigner A. MicroRNA replacement therapy for miR-145 and miR-33a
is efficacious in a model of colon carcinoma. Cancer Res. 2011;71(15):
5214–5224.
95. Ren D, Wang M, Guo W, et al. Wild-type p53 suppresses the epithelial-
mesenchymal transition and stemness in PC-3 prostate cancer cells by
modulating miR145. Int J Oncol. 2013;42(4):1473–1481.
96. Kent OA, Chivukula RR, Mullendore M, et al. Repression of the
miR-143/145 cluster by oncogenic Ras initiates a tumor-promoting
feed-forward pathway. Genes Dev. 2010;24(24):2754–2759.
97. Xu N, Papagiannakopoulos T, Pan G, Thomson Ja, Kosik KS.
MicroRNA-145 regulates OCT4, SOX2, and KLF4 and represses
pluripotency in human embryonic stem cells. Cell. 2009;137(4):
647–658.
98. Krizhanovsky V, Lowe SW. Stem cells: The promises and perils of
p53. Nature. 2009;460(7259):1085–1086.
99. Zhang J, Sun Q, Zhang Z, Ge S, Han ZG, Chen WT. Loss of microRNA-
143/145 disturbs cellular growth and apoptosis of human epithelial
cancers by impairing the MDM2-p53 feedback loop. Oncogene.
2013;32(1):61–69.
100. Pichiorri F, Suh S-s, Rocci A, et al. Downregulation of p53-inducible
microRNAs 192, 194,and 215 Impairs the p53/MDM2 Auto-
regulatory Loop in Multiple Myeloma Development. Cancer Cell.
2010;18(4):367–381.
101. Li S, Gao J, Gu J, Yuan J, Hua D, Shen L. MicroRNA-215 inhibits
relapse of colorectal cancer patients following radical surgery. Med
Oncol. 2013;30(2):549.
102. Georges SA, Biery MC, Kim SY, et al. Coordinated regulation of cell
cycle transcripts by p53-Inducible microRNAs, miR-192 and miR-215.
Cancer Res. 2008;68(24):10105–10112.
103. Geng L, Chaudhuri A, Talmon G, et al. MicroRNA-192 suppresses liver
metastasis of colon cancer. Oncogene. Epub November 11, 2013.
104. Dong P, Kaneuchi M, Watari H, et al. MicroRNA-194 inhibits epithelial
to mesenchymal transition of endometrial cancer cells by targeting
oncogene BMI-1. Mol Cancer. 2011;10:99.
105. Wellner U, Schubert J, Burk UC, et al. The EMT-activator ZEB1 pro-
motes tumorigenicity by repressing stemness-inhibiting microRNAs.
Nat Cell Biol. 2009;11(12):1487–1495.
106. Shimono Y, Zabala M, Cho RW, et al. Downregulation of miRNA-
200c links breast cancer stem cells with normal stem cells. Cell.
2009;138(3):592–603.
107. Lu YX, Yuan L, Xue XL, et al. Regulation of colorectal
carcinoma stemness, growth and metastasis by a miR-200-
c-Sox2 negative feedback loop mechanism. Clin Cancer Res.
2014;20(10):2631–2642.
108. Tanaka K, Miyata H, Yamasaki M, et al. Circulating miR-200c levels
significantly predict response to chemotherapy and prognosis of
patients undergoing neoadjuvant chemotherapy for esophageal cancer.
Ann Surg Oncol. 2013;20 Suppl 3:S607–S615.
109. Toiyama Y, Hur K, Tanaka K, et al. Serum miR-200c Is a Novel
Prognostic and Metastasis-Predictive Biomarker in Patients With
Colorectal Cancer. Ann Surg. 2014;259(4):735–743.
110. Valladares-Ayerbes M, Reboredo M, Medina-Villaamil V, et al. Circu-
lating miR-200c as a diagnostic and prognostic biomarker for gastric
cancer. J Transl Med. 2012;10:186.
111. Yu H, Duan B, Jiang L, et al. Serum miR-200c and clinical outcome
of patients with advanced esophageal squamous cancer receiving
platinum-based chemotherapy. Am J Transl Res. 2013;6(1):71–77.
112. Zhang GJ, Zhou T, Liu ZL, Tian HP, Xia SS. Plasma miR-200c and
miR-18a as potential biomarkers for the detection of colorectal
carcinoma. Mol Clin Oncol. 2013;1(2):379–384.
113. Tsai JH, Donaher JL, Murphy DA, Chau S, Yang J. Spatiotemporal reg-
ulation of epithelial-mesenchymal transition is essential for squamous
cell carcinoma metastasis. Cancer Cell. 2012;22(6):725–736.
114. Yamakuchi M, Lotterman CD, Bao C, et al. P53-induced microRNA-107
inhibits HIF-1 and tumor angiogenesis. Proc Natl Acad Sci U S A.
2010;107(14):6334–6339.
Clinical and Experimental Gastroenterology 2014:7
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
410
Rokavec et al
115. Martello G, Rosato A, Ferrari F, et al. A MicroRNA targeting dicer
for metastasis control. Cell. 2010;141(7):1195–1207.
116. Chen PS, Su JL, Cha ST, et al. miR-107 promotes tumor progression
by targeting the let-7 microRNA in mice and humans. J Clin Invest.
2011;121(9):3442–3455.
117. Chen HY, Lin YM, Chung HC, et al. miR-103/107 promote metastasis
of colorectal cancer by targeting the metastasis suppressors DAPK and
KLF4. Cancer Res. 2012;72(14):3631–3641.
118. Li X, Zhang Y, Shi Y, et al. MicroRNA-107, an oncogene microRNA
that regulates tumour invasion and metastasis by targeting DICER1
in gastric cancer. J Cell Mol Med. 2011;15(9):1887–1895.
119. Inoue T, Iinuma H, Ogawa E, Inaba T, Fukushima R. Clinicopathological
and prognostic significance of microRNA-107 and its relationship to
DICER1 mRNA expression in gastric cancer. Oncol Rep. 2012;27(6):
1759–1764.
120. Feng L, Xie Y, Zhang H, Wu Y. miR-107 targets cyclin-dependent
kinase 6 expression, induces cell cycle G1 arrest and inhibits invasion
in gastric cancer cells. Med Oncol. 2012;29(2):856–863.
121. Kumar MS, Lu J, Mercer KL, Golub TR, Jacks T. Impaired microRNA
processing enhances cellular transformation and tumorigenesis.
Nat Genet. 2007;39(5):673–677.
122. Li F, Liu B, Gao Y, et al. Upregulation of MicroRNA-107 induces
proliferation in human gastric cancer cells by targeting the transcrip-
tion factor FOXO1. FEBS Lett. 2014;588(4):538–544.
123. Chen L, Chen XR, Chen FF, et al. MicroRNA-107 inhibits U87 glioma
stem cells growth and invasion. Cell Mol Neurobiol. 2013;33(5):
651–657.
124. Liang M, Yao G, Yin M, et al. Transcriptional cooperation between p53
and NF-κB p65 regulates microRNA-224 transcription in mouse ovar-
ian granulosa cells. Mol Cell Endocrinol. 2013;370(1–2):119–129.
125. Zhai H, Song B, Xu X, Zhu W, Ju J. Inhibition of autophagy and
tumor growth in colon cancer by miR-502. Oncogene. 2013;32(12):
1570–1579.
126. Yan HL, Xue G, Mei Q, et al. Repression of the miR-17-92 cluster
by p53 has an important function in hypoxia-induced apoptosis.
EMBO J. 2009;28(18):2719–2732.
127. Tsuchida A, Ohno S, Wu W, et al. miR-92 is a key oncogenic
component of the miR-17-92 cluster in colon cancer. Cancer Sci.
2011;102(12):2264–2271.
128. Olive V, Jiang I, He L. mir-17-92, a cluster of miRNAs in the midst of the
cancer network. Int J Biochem Cell Biol. 2010;42(8):1348–1354.
129. Le MT, Teh C, Shyh-Chang N, et al. MicroRNA-125b is a novel
negative regulator of p53. Genes Dev. 2009;23(7):862–876.
130. Le MT, Shyh-Chang N, Khaw SL, et al. Conserved regulation of p53
network dosage by microRNA-125b occurs through evolving miRNA-
target gene pairs. PLoS Genet. 2011;7(9):e1002242.
131. Giray BG, Emekdas G, Tezcan S, et al. Profiles of serum microRNAs;
miR-125b-5p and miR223-3p serve as novel biomarkers for HBV-
positive hepatocellular carcinoma. Mol Biol Rep. 2014;41(7):
4513–4519.
132. Nishida N, Yokobori T, Mimori K, et al. MicroRNA miR-125b is a
prognostic marker in human colorectal cancer. Int J Oncol. 2011;38(5):
1437–1443.
133. Hu W, Chan CS, Wu R, et al. Negative regulation of tumor suppressor
p53 by microRNA miR-504. Mol Cell. 2010;38(5):689–699.
134. Kumar M, Lu Z, Takwi AA, et al. Negative regulation of the tumor
suppressor p53 gene by microRNAs. Oncogene. 2011;30(7):
843–853.
135. Herrera-Merchan A, Cerrato C, Luengo G, et al. miR-33-mediated
downregulation of p53 controls hematopoietic stem cell self-renewal.
Cell Cycle. 2010;9(16):3277–3285.
136. Swarbrick A, Woods SL, Shaw A, et al. miR-380-5p represses p53 to
control cellular survival and is associated with poor outcome in MYCN-
amplified neuroblastoma. Nat Med. 2010;16(10):1134–1140.
137. Xu X, Chen Z, Zhao X, et al. MicroRNA-25 promotes cell migration
and invasion in esophageal squamous cell carcinoma. Biochem Biophys
Res Commun. 2012;421(4):640–645.
138. Zhang N, Wei X, Xu L. miR-150 promotes the proliferation of lung
cancer cells by targeting P53. FEBS Lett. 2013;587(15):2346–2351.
139. Liu Y, Xing R, Zhang X, et al. miR-375 targets the p53 gene to regulate
cellular response to ionizing radiation and etoposide in gastric cancer
cells. DNA Repair (Amst). 2013;12(9):741–750.
140. Tian S, Huang S, Wu S, Guo W, Li J, He X. MicroRNA-1285 inhibits
the expression of p53 by directly targeting its 3 untranslated region.
Biochem Biophys Res Commun. 2010;396(2):435–439.
141. Xu CX, Xu M, Tan L, et al. MicroRNA miR-214 regulates ovarian can-
cer cell stemness by targeting p53/Nanog. J Biol Chem. 2012;287(42):
34970–34978.
142. Zhang Y, Gao JS, Tang X, et al. MicroRNA 125a and its
regulation of the p53 tumor suppressor gene. FEBS Lett.
2009;583(22):3725–3730.
143. Zhang S, Zhang C, Li Y, Wang P, Yue Z, Xie S. miR-98 regulates
cisplatin-induced A549 cell death by inhibiting TP53 pathway. Biomed
Pharmacother. 2011;65(6):436–442.
144. Wang DT, Ma ZL, Li YL, et al. miR-150, p53 protein and relevant
miRNAs consist of a regulatory network in NSCLC tumorigenesis.
Oncol Rep. 2013;30(1):492–498.
145. Wu C, Li M, Hu C, Duan H. Clinical significance of serum miR-223,
miR-25 and miR-375 in patients with esophageal squamous cell
carcinoma. Mol Biol Rep. 2014;41(3):1257–1266.
146. Li X, Yang C, Wang X, Zhang J, Zhang R, Liu R. The expression of
miR-25 is increased in colorectal cancer and is associated with patient
prognosis. Med Oncol. 2014;31(1):781.
147. Yang X, Zhong X, Tanyi JL, et al. mir-30d Regulates multiple
genes in the autophagy pathway and impairs autophagy process in
human cancer cells. Biochem Biophys Res Commun. 2013;431(3):
617–622.
148. Li N, Kaur S, Greshock J, et al. A combined array-based comparative
genomic hybridization and functional library screening approach
identifies mir-30d as an oncomir in cancer. Cancer Res. 2012;72(1):
154–164.
149. Ugalde AP, Ramsay AJ, de la Rosa J, et al. Aging and chronic DNA
damage response activate a regulatory pathway involving miR-29 and
p53. EMBO J. 2011;30(11):2219–2232.
150. Park SY, Lee JH, Ha M, Nam JW, Kim VN. miR-29 miRNAs acti-
vate p53 by targeting p85 alpha and CDC42. Nat Struct Mol Biol.
2009;16(1):23–29.
151. Wang Y, Zhang X, Li H, Yu J, Ren X. The role of miRNA-29 family
in cancer. Eur J Cell Biol. 2013;92(3):123–128.
152. Gong J, Li J, Wang Y, et al. Characterization of microRNA-29 family
expression and investigation of their mechanistic roles in gastric cancer.
Carcinogenesis. 2014;35(2):497–506.
153. Zhu H-T, Dong Q-Z, Sheng Y-Y, et al. MicroRNA-29a-5p is a novel
predictor for early recurrence of hepatitis B virus-related hepatocellular
carcinoma after surgical resection. PLoS One. 2012;7(12):e52393.
154. Xiong Y, Fang JH, Yun JP, et al. Effects of microRNA-29 on
apoptosis, tumorigenicity, and prognosis of hepatocellular carcinoma.
Hepatology. 2010;51(3):836–845.
155. Gramantieri L, Fornari F, Callegari E, et al. MicroRNA involve-
ment in hepatocellular carcinoma. J Cell Mol Med. 2008;12(6A):
2189–2204.
156. Fornari F, Gramantieri L, Giovannini C, et al. MiR-122/cyclin G1
interaction modulates p53 activity and affects doxorubicin sensi-
tivity of human hepatocarcinoma cells. Cancer Res. 2009;69(14):
5761–5767.
157. Okamoto K, Li H, Jensen MR, et al. Cyclin G recruits PP2A to
dephosphorylate Mdm2. Mol Cell. 2002;9(4):761–771.
158. Okamoto K, Beach D. Cyclin G is a transcriptional target of the p53
tumor suppressor protein. EMBO J. 1994;13(20):4816–4822.
159. Hünten S, Siemens H, Kaller M, Hermeking H. The p53/microRNA
network in cancer: experimental and bioinformatics approaches. Adv
Exp Med Biol. 2013;774:77–101.
160. Concepcion CP, Han YC, Mu P, et al. Intact p53-dependent responses
in miR-34-deficient mice. PLoS Genet. 2012;8(7):e1002797.
Clinical and Experimental Gastroenterology 2014:7 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
411
The p53/microRNA axis in GI cancer
161. Xue W, Dahlman JE, Tammela T, et al. Small RNA combination therapy for
lung cancer. Proc Natl Acad Sci USA. 2014;111(34):E3553–3561.
162. Nugent M, Miller N, Kerin MJ. Circulating miR-34a levels are reduced
in colorectal cancer. J Surg Oncol. 2012;106(8):947–952.
163. Wu J, Wu G, Lv L, et al. MicroRNA-34a inhibits migration and
invasion of colon cancer cells via targeting to Fra-1. Carcinogenesis.
2012;33(3):519–528.
164. Chen X, Hu H, Guan X, et al. CpG island methylation status of
miRNAs in esophageal squamous cell carcinoma. Int J Cancer.
2012;130(7):1607–1613.
165. Jamieson NB, Morran DC, Morton JP, et al. MicroRNA molecular
prof iles associated with diagnosis, clinicopathologic criteria,
and overall survival in patients with resectable pancreatic ductal
adenocarcinoma. Clin Cancer Res. 2012;18(2):534–545.
166. Harata K, Ishiguro H, Kuwabara Y, Kimura M, Mitsui A. MicroRNA-34b
has an oncogenic role in esophageal squamous cell carcinoma. Oncol
Lett. 2010;1(4):685–689.
167. Liu SG, Qin XG, Zhao BS, et al. Differential expression of miRNAs
in esophageal cancer tissue. Oncol Lett. 2013;5(5):1639–1642.
168. Diaz T, Tejero R, Moreno I, et al. Role of miR-200 family members in
survival of colorectal cancer patients treated with fluoropyrimidines.
J Surg Oncol. 2014;109(7):676–683.
169. Hur K, Toiyama Y, Takahashi M, et al. MicroRNA-200c modulates
epithelial-to-mesenchymal transition (EMT) in human colorectal
cancer metastasis. Gut. 2013;62(9):1315–1326.
170. Chen J, Wang W, Zhang Y, Chen Y, Hu T. Predicting distant metastasis and
chemoresistance using plasma miRNAs. Med Oncol. 2014; 31(1):799.
171. Davalos V, Moutinho C, Villanueva A, et al. Dynamic epigenetic regulation
of the microRNA-200 family mediates epithelial and mesenchymal transi-
tions in human tumorigenesis. Oncogene. 2012;31(16): 2062–2074.
172. Du Y, Xu Y, Ding L, et al. Down-regulation of miR-141 in gastric
cancer and its involvement in cell growth. J Gastroenterol. 2009;44(6):
556–561.
173. Karakatsanis A, Papaconstantinou I, Gazouli M, Lyberopoulou A,
Polymeneas G, Voros D. Expression of microRNAs, miR-21, miR-31,
miR-122, miR-145, miR-146a, miR-200c, miR-221, miR-222, and
miR-223 in patients with hepatocellular carcinoma or intrahepatic
cholangiocarcinoma and its prognostic significance. Mol Carcinog.
2013;52(4):297–303.
174. Yeh TS, Wang F, Chen TC, et al. Expression profile of microRNA-200
family in hepatocellular carcinoma with bile duct tumor thrombus.
Ann Surg. 2014;259(2):346–354.
175. Sun Y, Shen S, Liu X, et al. MiR-429 inhibits cells growth and inva-
sion and regulates EMT-related marker genes by targeting Onecut2
in colorectal carcinoma. Mol Cell Biochem. 2014;390(1-2):19–30.
176. Chen Z, Saad R, Jia P, et al. Gastric adenocarcinoma has a unique
microRNA signature not present in esophageal adenocarcinoma.
Cancer. 2013;119(11):1985–1993.
177. Li A, Omura N, Hong SM, et al. Pancreatic cancers epigenetically
silence SIP1 and hypomethylate and overexpress miR-200a/200b in
association with elevated circulating miR-200a and miR-200b levels.
Cancer Res. 2010;70(13):5226–5237.
178. Sharma P, Saraya A, Gupta P, Sharma R. Decreased levels of circu-
lating and tissue miR-107 in human esophageal cancer. Biomarkers.
2013;18(4):322–330.
179. Lee KH, Lotterman C, Karikari C, et al. Epigenetic silencing of
MicroRNA miR-107 regulates cyclin-dependent kinase 6 expression
in pancreatic cancer. Pancreatology. 2009;9(3):293–301.
180. Feng Y, Zhu J, Ou C, et al. MicroRNA-145 inhibits tumour growth
and metastasis in colorectal cancer by targeting fascin-1. Br J Cancer.
2014;110(9):2300–2309.
181. Earle JS, Luthra R, Romans A, et al. Association of microRNA expres-
sion with microsatellite instability status in colorectal adenocarcinoma.
J Mol Diagn. 2010;12(4):433–440.
182. Li JM, Zhao RH, Li ST, et al. Down-regulation of fecal miR-143 and
miR-145 as potential markers for colorectal cancer. Saudi Med J.
2012;33(1):24–29.
183. Qiu T, Zhou X, Wang J, et al. MiR-145, miR-133a and miR-133b
inhibit proliferation, migration, invasion and cell cycle progression
via targeting transcription factor Sp1 in gastric cancer. FEBS Lett.
2014;588(7):1168–1177.
184. Xing AY, Wang B, Shi DB, et al. Deregulated expression of miR-145
in manifold human cancer cells. Exp Mol Pathol. 2013;95(1):91–97.
185. Chiang Y, Song Y, Wang Z, et al. microRNA-192, -194 and -215
are frequently downregulated in colorectal cancer. Exp Ther Med.
2012;3(3):560–566.
186. Faltejskova P, Svoboda M, Srutova K, et al. Identification and functional
screening of microRNAs highly deregulated in colorectal cancer.
J Cell Mol Med. 2012;16(11):2655–2666.
187. Kahlert C, Klupp F, Brand K, et al. Invasion front-specific expression
and prognostic significance of microRNA in colorectal liver metastases.
Cancer Sci. 2011;102(10):1799–1807.
188. Song Y, Zhao F, Wang Z, et al. Inverse association between miR-194
expression and tumor invasion in gastric cancer. Ann Surg Oncol.
2012;19 Suppl 3:S509–S517.
189. Deng Y, Huang Z, Xu Y, et al. MiR-215 modulates gastric cancer cell
proliferation by targeting RB1. Cancer Lett. 2014;342(1):27–35.
190. Wang LG, Gu J. Serum microRNA-29a is a promising novel marker
for early detection of colorectal liver metastasis. Cancer Epidemiol.
2012;36(1):e61–e67.
191. Tang W, Zhu Y, Gao J, et al. MicroRNA-29a promotes colorectal cancer
metastasis by regulating matrix metalloproteinase 2 and E-cadherin
via KLF4. Br J Cancer. 2014;110(2):450–458.
192. Chen L, Xiao H, Wang Z-H, et al. miR-29a suppresses growth and
invasion of gastric cancer cells in vitro by targeting VEGF-A. BMB
Rep. 2014;47(1):39–44.
193. Xiao J, Lin H, Luo X, Luo X, Wang Z. miR-605 joins p53 network
to form a p53:miR-605:Mdm2 positive feedback loop in response to
stress. EMBO J. 2011;30(3):524–532.
194. Jin L, Hu WL, Jiang CC, et al. MicroRNA-149*, a p53-responsive
microRNA, functions as an oncogenic regulator in human melanoma.
Proc Natl Acad Sci U S A. 2011;108(38):15840–15845.
195. Øster B, Linnet L, Christensen LL, et al; COLOFOL steering
group. Non-CpG island promoter hypomethylation and miR-149
regulate the expression of SRPX2 in colorectal cancer. Int J Cancer.
2013;132(10):2303–2315.
196. Wang F, Ma YL, Zhang P, et al. SP1 mediates the link between methy-
lation of the tumour suppressor miR-149 and outcome in colorectal
cancer. J Pathol. 2013;229(1):12–24.
197. Wang Y, Zheng X, Zhang Z, et al. MicroRNA-149 inhibits proliferation
and cell cycle progression through the targeting of ZBTB2 in human
gastric cancer. PLoS One. 2012;7(10):e41693.
198. Lin J, Huo R, Xiao L, et al. A novel p53/microRNA-22/Cyr61 axis in
synovial cells regulates inflammation in rheumatoid arthritis. Arthritis
Rheumatol. 2014;66(1):49–59.
199. Zhang G, Xia S, Tian H, Liu Z, Zhou T. Clinical significance of
miR-22 expression in patients with colorectal cancer. Med Oncol.
2012;29(5):3108–3112.
200. Zhang C, Wang C, Chen X, et al. Expression profile of microRNAs
in serum: a fingerprint for esophageal squamous cell carcinoma. Clin
Chem. 2010;56(12):1871–1879.
201. Ganepola GA, Rutledge JR, Suman P, Yiengpruksawan A, Chang
DH. Novel blood-based microRNA biomarker panel for early
diagnosis of pancreatic cancer. World J Gastrointest Oncol.
2014;6(1):22–33.
202. Guo MM, Hu LH, Wang YQ, et al. miR-22 is down-regulated in gastric
cancer, and its overexpression inhibits cell migration and invasion via
targeting transcription factor Sp1. Med Oncol. 2013;30(2):542.
203. Wang W, Li F, Zhang Y, Tu Y, Yang Q, Gao X. Reduced expression of
miR-22 in gastric cancer is related to clinicopathologic characteristics
or patient prognosis. Diagn Pathol. 2013;8:102.
204. Zhou L, He J, Zhang Y. MicroRNA-22 expression in hepatocellular
carcinoma and its correlation with ezrin protein. J Int Med Res.
2013;41(4):1009–1016.
Clinical and Experimental Gastroenterology 2014:7
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
412
Rokavec et al
205. Zhang J, Yang Y, Yang T, et al. microRNA-22, downregulated in
hepatocellular carcinoma and correlated with prognosis, suppresses
cell proliferation and tumourigenicity. Br J Cancer. 2010;103(8):
1215–1220.
206. Au Yeung CL, Tsang TY, Yau PL, Kwok TT. Human papillomavirus
type 16 E6 induces cervical cancer cell migration through the p53/
microRNA-23b/urokinase-type plasminogen activator pathway. Onco-
gene. 2011;30(21):2401–2410.
207. Zhao BS, Liu SG, Wang TY, et al. Screening of microRNA in
patients with esophageal cancer at same tumor node metastasis
stage with different prognoses. Asian Pac J Cancer Prev. 2013;
14(1): 139–143.
208. Salvi A, Sabelli C, Moncini S, et al. MicroRNA-23b mediates uroki-
nase and c-met downmodulation and a decreased migration of human
hepatocellular carcinoma cells. FEBS J. 2009;276(11):2966–2982.
209. Zhang H, Hao Y, Yang J, et al. Genome-wide functional screening of
miR-23b as a pleiotropic modulator suppressing cancer metastasis.
Nat Commun. 2011;2:554.
210. Piovan C, Palmieri D, Di Leva G, et al. Oncosuppressive role of
p53-induced miR-205 in triple negative breast cancer. Mol Oncol.
2012;6(4):458–472.
211. Gu J, Wang Y, Wu X. MicroRNA in the pathogenesis and prognosis
of esophageal cancer. Curr Pharm Des. 2013;19(7):1292–1300.
212. Saad R, Chen Z, Zhu S, et al. Deciphering the unique microRNA sig-
nature in human esophageal adenocarcinoma. PLoS One. 2013;8(5):
e64463.
213. Zhang T, Zhang J, Cui M, et al. Hepatitis B virus X protein inhibits
tumor suppressor miR-205 through inducing hypermethylation of
miR-205 promoter to enhance carcinogenesis. Neoplasia. 2013;15(11):
1282–1291.
214. Zhang Y, Liao JM, Zeng SX, Lu H. p53 downregulates Down
syndrome-associated DYRK1A through miR-1246. EMBO Rep.
2011;12(8):811–817.
215. Piepoli A, Tavano F, Copetti M, et al. Mirna expression profiles
identify drivers in colorectal and pancreatic cancers. PLoS One.
2012;7(3):e33663.
216. Ogata-Kawata H, Izumiya M, Kurioka D, et al. Circulating exosomal
microRNAs as biomarkers of colon cancer. PLoS One. 2014;9(4):
e92921.
217. Fu HL, Wu de P, Wang XF, et al. Altered miRNA expression is asso-
ciated with differentiation, invasion, and metastasis of esophageal
squamous cell carcinoma (ESCC) in patients from Huaian, China.
Cell Biochem Biophys. 2013;67(2):657–668.
218. Takeshita N, Hoshino I, Mori M, et al. Serum microRNA expression
profile: miR-1246 as a novel diagnostic and prognostic biomarker for
oesophageal squamous cell carcinoma. Br J Cancer. 2013;108(3):
644–652.
219. Barsotti AM, Beckerman R, Laptenko O, Huppi K, Caplen NJ,
Prives C. p53-Dependent induction of PVT1 and miR-1204. J Biol
Chem. 2012;287(4):2509–2519.
220. Koga Y, Yasunaga M, Takahashi A, et al. MicroRNA expression profil-
ing of exfoliated colonocytes isolated from feces for colorectal cancer
screening. Cancer Prev Res (Phila). 2010;3(11):1435–1442.
221. Diosdado B, van de Wiel MA, Terhaar Sive Droste JS, et al.
MiR-17-92 cluster is associated with 13q gain and c-myc expression
during colorectal adenoma to adenocarcinoma progression. Br J
Cancer. 2009;101(4):707–714.
222. Xu XL, Jiang YH, Feng JG, Su D, Chen PC, Mao WM. MicroRNA-17,
microRNA-18a, and microRNA-19a are prognostic indicators in
esophageal squamous cell carcinoma. Ann Thorac Surg. 2014;97(3):
1037–1045.
223. Liu M, Wang Z, Yang S, et al. TNF-alpha is a novel target of miR-19a.
Int J Oncol. 2011;38(4):1013–1022.
224. Tsujiura M, Komatsu S, Ichikawa D, et al. Circulating miR-18a in
plasma contributes to cancer detection and monitoring in patients with
gastric cancer. Gastric Cancer. Epub March 14, 2014.
225. Shiotani A, Uedo N, Iishi H, et al. H. pylori eradication did not improve
dysregulation of specific oncogenic miRNAs in intestinal metaplastic
glands. J Gastroenterol. 2012;47(9):988–998.
226. Shigoka M, Tsuchida A, Matsudo T, et al. Deregulation of miR-92a
expression is implicated in hepatocellular carcinoma development.
Pathol Int. 2010;60(5):351–357.
227. Morimura R, Komatsu S, Ichikawa D, et al. Novel diagnostic value
of circulating miR-18a in plasma of patients with pancreatic cancer.
Br J Cancer. 2011;105(11):1733–1740.
228. Zhang GJ, Zhou H, Xiao HX, Li Y, Zhou T. Up-regulation of miR-224
promotes cancer cell proliferation and invasion and predicts relapse
of colorectal cancer. Cancer Cell Int. 2013;13(1):104.
229. Liao WT, Li TT, Wang ZG, et al. microRNA-224 promotes cell
proliferation and tumor growth in human colorectal cancer by
repressing PHLPP1 and PHLPP2. Clin Cancer Res. 2013;19(17):
4662–4672.
230. Yuan K, Xie K, Fox J, et al. Decreased levels of miR-224 and the
passenger strand of miR-221 increase MBD2, suppressing maspin
and promoting colorectal tumor growth and metastasis in mice.
Gastroenterology. 2013;145(4):853–864. e9.
231. Scisciani C, Vossio S, Guerrieri F, et al. Transcriptional regulation of
miR-224 upregulated in human HCCs by NFkappaB inflammatory
pathways. J Hepatol. 2012;56(4):855–861.
232. Zhang Y, Takahashi S, Tasaka A, Yoshima T, Ochi H, Chayama K.
Involvement of microRNA-224 in cell proliferation, migration, inva-
sion, and anti-apoptosis in hepatocellular carcinoma. J Gastroenterol
Hepatol. 2013;28(3):565–575.
233. Komatsu S, Ichikawa D, Hirajima S, et al. Plasma microRNA profiles:
identification of miR-25 as a novel diagnostic and monitoring biomarker
in oesophageal squamous cell carcinoma. Br J Cancer. 2014.
234. Zhu C, Ren C, Han J, et al. A five-microRNA panel in plasma was
identified as potential biomarker for early detection of gastric cancer.
Br J Cancer. 2014;110(9):2291–2299.
235. Zhao H, Wang Y, Yang L, Jiang R, Li W. MiR-25 promotes gastric
cancer cells growth and motility by targeting RECK. Mol Cell
Biochem. 2014;385(1–2):207–213.
236. Ueda T, Volinia S, Okumura H, et al. Relation between microRNA
expression and progression and prognosis of gastric cancer:
a microRNA expression analysis. Lancet Oncol. 2010;11(2):136–146.
237. Kim BH, Hong SW, Kim A, Choi SH, Yoon SO. Prognostic implica-
tions for high expression of oncogenic microRNAs in advanced gastric
carcinoma. J Surg Oncol. 2013;107(5):505–510.
238. Yao J, Liang L, Huang S, et al. MicroRNA-30d promotes tumor inva-
sion and metastasis by targeting Galphai2 in hepatocellular carcinoma.
Hepatology. 2010;51(3):846–856.
239. Huang S-D, Yuan Y, Zhuang C-W, et al. MicroRNA-98 and microRNA-
214 post-transcriptionally regulate enhancer of zeste homolog 2 and
inhibit migration and invasion in human esophageal squamous cell
carcinoma. Mol Cancer. 2012;11:51.
240. Fassan M, Pizzi M, Realdon S, et al. The HER2-miR125a5p/
miR125b loop in gastric and esophageal carcinogenesis. Hum Pathol.
2013;44(9):1804–1810.
241. Nishida N, Mimori K, Fabbri M, et al. MicroRNA-125a-5p is an inde-
pendent prognostic factor in gastric cancer and inhibits the prolifera-
tion of human gastric cancer cells in combination with trastuzumab.
Clin Cancer Res. 2011;17(9):2725–2733.
242. Hashiguchi Y, Nishida N, Mimori K, et al. Down-regulation of
miR-125a-3p in human gastric cancer and its clinicopathological
significance. Int J Oncol. 2012;40(5):1477–1482.
243. Bi Q, Tang S, Xia L, et al. Ectopic expression of MiR-125a inhibits the
proliferation and metastasis of hepatocellular carcinoma by targeting
MMP11 and VEGF. PLoS One. 2012;7(6):e40169.
244. Kim JK, Noh JH, Jung KH, et al. Sirtuin7 oncogenic potential in
human hepatocellular carcinoma and its regulation by the tumor
suppressors MiR-125a-5p and MiR-125b. Hepatology. 2013;57(3):
1055–1067.
Clinical and Experimental Gastroenterology
Publish your work in this journal
Submit your manuscript here: http://www.dovepress.com/clinical-and-experimental-gastroenterology-journal
Clinical and Experimental Gastroenterology is an international, peer-
reviewed, open access journal, publishing all aspects of gastroenterology
in the clinic and laboratory, including: Pathology, pathophysiology
of gastrointestinal disease; Investigation and treatment of gastointes-
tinal disease; Pharmacology of drugs used in the alimentary tract;
Immunology/genetics/genomics related to gastrointestinal disease.
This journal is indexed on CAS. The manuscript management system
is completely online and includes a very quick and fair peer-review
system. Visit http://www.dovepress.com/testimonials.php to read real
quotes from published authors.
Clinical and Experimental Gastroenterology 2014:7 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
Dovepress
413
The p53/microRNA axis in GI cancer
245. Liang L, Wong C-M, Ying Q, et al. MicroRNA-125b suppressesed
human liver cancer cell proliferation and metastasis by directly target-
ing oncogene LIN28B2. Hepatology. 2010;52(5):1731–1740.
246. Bloomston M, Frankel WL, Petrocca F, et al. MicroRNA expression
patterns to differentiate pancreatic adenocarcinoma from normal
pancreas and chronic pancreatitis. JAMA. 2007;297(17):1901–1908.
247. Ma Y, Zhang P, Wang F, et al. miR-150 as a potential biomarker asso-
ciated with prognosis and therapeutic outcome in colorectal cancer.
Gut. 2012;61(10):1447–1453.
248. Yokobori T, Suzuki S, Tanaka N, et al. MiR-150 is associated with
poor prognosis in esophageal squamous cell carcinoma via targeting
the EMT inducer ZEB1. Cancer Sci. 2013;104(1):48–54.
249. Wu Q, Jin H, Yang Z, et al. MiR-150 promotes gastric cancer prolifera-
tion by negatively regulating the pro-apoptotic gene EGR2. Biochem
Biophys Res Commun. 2010;392(3):340–345.
250. Srivastava SK, Bhardwaj A, Singh S, et al. MicroRNA-150 directly
targets MUC4 and suppresses growth and malignant behavior of
pancreatic cancer cells. Carcinogenesis. 2011;32(12):1832–1839.
251. Zhou Y, Hong L. Prediction value of miR-483 and miR-214 in progno-
sis and multidrug resistance of esophageal squamous cell carcinoma.
Genet Test Mol Biomarkers. 2013;17(6):470–474.
252. Yang TS, Yang XH, Wang XD, Wang YL, Zhou B, Song ZS. MiR-214
regulate gastric cancer cell proliferation, migration and invasion by
targeting PTEN. Cancer Cell Int. 2013;13(1):68.
253. Shih TC, Tien YJ, Wen CJ, et al. MicroRNA-214 downregulation
contributes to tumor angiogenesis by inducing secretion of the
hepatoma-derived growth factor in human hepatoma. J Hepatol.
2012;57(3):584–591.
254. Xia H, Ooi LL, Hui KM. MiR-214 targets β-catenin pathway to sup-
press invasion, stem-like traits and recurrence of human hepatocellular
carcinoma. PLoS One. 2012;7(9):e44206.
255. Wang J, Li J, Wang X, Zheng C, Ma W. Downregulation of microRNA-
214 and overexpression of FGFR-1 contribute to hepatocellular carci-
noma metastasis. Biochem Biophys Res Commun. 2013;439(1):47–53.
... In addition, the p53 protein regulates miRNA processing, primarily through its central DNA-binding domain, a target site of most cancerspecific mutations. Interestingly, miRNAs can also regulate p53 expression by matching in the seed region into the 3′UTR of TP53 mRNA, directly inducing the repression of TP53 or its regulators (Hermeking, 2012;Hermeking et al., 2014). Although several tumor-specific alterations in the p53-miRNA network have been described in different cancers (Hermeking, 2012;Datta et al., 2019), there is no data on miRNAs targeting of TP53 gene in HPV-associated penile cancer. ...
... More than 20 miRNAs have been described to directly regulate p53 via canonical bindings (seed) in 3′UTR (reviewed by Liu et al., 2017). Down regulation of TP53 through seed sequences induce phenotypes that are consistent with loss of p53 function, such as reduced apoptosis, cellular senescence, increased invasion, and growth of tumor cells (Hermeking, 2012;Deng and Sui, 2013;Hermeking et al., 2014). Despite the increasing number of miRNAs that form the TP53 mRNA/miRNAs interaction network, there is no information on TP53-repressor miRNAs in HPV-associated PeCa. ...
Article
Full-text available
Cancer development by the human papillomavirus (HPV) infection can occur through the canonical HPV/p53/RB1 pathway mediated by the E2/E6/E7 viral oncoproteins. During the transformation process, HPV inserts its genetic material into host Integration Sites (IS), affecting coding genes and miRNAs. In penile cancer (PeCa) there is limited data on the miRNAs that regulate mRNA targets associated with HPV, such as the TP53 and RB1 genes. Considering the high frequency of HPV infection in PeCa patients in Northeast Brazil, global miRNA expression profiling was performed in high-risk HPV-associated PeCa that presented with TP53 and RB1 mRNA downregulated expression. The miRNA expression profile of 22 PeCa tissue samples and five non-tumor penile tissues showed 507 differentially expressed miRNAs: 494 downregulated and 13 upregulated (let-7a-5p, miR-130a-3p, miR-142-3p, miR-15b-5p miR-16-5p, miR-200c-3p, miR-205-5p, miR-21-5p, miR-223-3p, miR-22-3p, miR-25-3p, miR-31-5p and miR-93-5p), of which 11 were identified to be in HPV16-IS and targeting TP53 and RB1 genes. One hundred and thirty-one and 490 miRNA binding sites were observed for TP53 and RB1, respectively, most of which were in seedless regions. These findings suggest that up-regulation of miRNA expression can directly repress TP53 and RB1 expression by their binding sites in the non-canonical seedless regions.
... 61 Contemporary scientific knowledge suggests that albeit P53 regulates transcription and maturation of multiple downstream tiny RNAs by activation or repression of distinct molecules, vice versa, TP53 expression is also under the tight control of particular microRNAs. 62 For instance, Shi et al reported that P53 can induce miR-15a/16-1 to form a double-negative feedback loop with transcription factor AP4. 63 Then, activated AP4 performs critical function regarding invasiveness and metastasis of colorectal malignancies. In addition, in silico searches identified miR-504 as a novel microRNA that can negatively regulate p53 expression via two binding sites in the human p53-3′UTR. ...
Article
Full-text available
Colorectal carcinoma (colon and rectum) is currently considered among the most prevalent malignancies of Western societies. The pathogenesis and etiological mechanisms underlying colorectal cancer (CRC) development remain complex and heterogeneous. The homeostasis and function of normal human intestinal cells is highly regulated by microRNAs. Therefore, it is not surprising that mutations and inactivation of these molecules appear to be linked with progression of colorectal tumors. Recent studies have reported significant alterations of microRNA expression in adenomas and CRCs compared with adjacent normal tissues. This observed deviation has been proposed to correlate with the progression and survival of disease as well as with choice of optimal treatment and drug resistance. MicroRNAs can adopt either oncogenic or tumor-suppressive roles during regulation of pathways that drive carcinogenesis. Typically, oncogenic microRNAs termed oncomirs, target and silence endogenous tumor-suppressor genes. On the other hand, tumor-suppressive microRNAs are critical in downregulating genes associated with cell growth and malignant capabilities. By extensively evaluating robust studies, we have emphasized and distinguished a discrete set of microRNAs that can modulate tumor progression by silencing specific driver genes crucial in signaling pathways including Wnt/b-catenin, epidermal growth factor receptor, P53, mismatch repair DNA repair, and transforming-growth factor beta.
... By referring to previous research findings, 31 OXA-or hypoxia-induced miRs were selected. Since numerous of these miRs are P53 dependent (Table I) (73)(74)(75)(76), they are probably involved in the regulation of resistance by P53. RT-qPCR results indicated that among these miRs, 6 miRs were induced by OXA, and 8miRs were induced by hypoxia. ...
Article
Full-text available
Oxaliplatin (OXA)‑containing regimens are used as first‑line chemotherapy in colorectal cancer (CRC). However, OXA resistance remains a major challenge in CRC treatment. CRC cells that adapt to hypoxia can potentially develop OXA resistance, and the underlying molecular mechanisms still need to be further investigated. In the current study, the OXA drug sensitivity of two CRC cell lines, HCT116 (TP53WT) and HT29 (TP53MT), was compared under both normoxic and hypoxic conditions. It was found that under normoxic condition, HCT116 cells showed significantly higher OXA sensitivity than HT29 cells. However, both cell lines showed remarkable OXA resistance under hypoxic conditions. It was also revealed that P53 levels were increased after OXA and hypoxia treatment in HCT116 cells but not in HT29 cells. Notably, knocking down P53WT decreased normoxic but increased hypoxic OXA sensitivity in HCT116 cells, which did not exist in HT29 cells. Molecular analysis indicated that P53WT activated microRNA (miR)‑26a and miR‑34a in OXA treatment and activated miR‑23a in hypoxia treatment. Cell proliferation experiments indicated that a high level of miR‑23a decreased OXA sensitivity and that a high level of miR‑26a or miR‑34a increased OXA sensitivity in HCT116 cells. Additionally, it was demonstrated that miR‑26a, miR‑34a and miR‑23a affect cell apoptosis through regulation of MCL‑1, EZH2, BCL‑2, SMAD 4 and STAT3. Taken together, the present findings revealed the dual function of P53 in regulating cellular chemo‑sensitivity and highlighted the role of P53‑miR interactions in the response of CRC cells to OXA chemotherapy under normoxic and hypoxic conditions.
... As a result, miRNAs can influence the expression of certain genes and take part in the regulation of a number of biological processes in the human organism [24]. Crucially, miRNAs are believed to influence the development of cardiovascular [6], oncological [42], and gastrointestinal diseases [30], as well as viral infections [35]. Because of this ability, the study of miRNAs and their respective targets may prove crucial for the design of novel diagnostic and treatment methods. ...
Preprint
Full-text available
Background: MicroRNAs (miRNAs) are short, non-coding RNA molecules that regulate gene expression by binding to specific mRNAs, inhibiting their translation. They play a critical role in regulating various biological processes and are implicated in many diseases, including cardiovascular, oncological, gastrointestinal diseases, and viral infections. Computational methods that can identify potential miRNA-mRNA interactions from raw data use one-dimensional miRNA-mRNA duplex representations and simple sequence encoding techniques, which may limit their performance. Results: We have developed GraphTar, a new target prediction method that uses a novel graph-based representation to reflect the spatial structure of the miRNA-mRNA duplex. Unlike existing approaches, we use the word2vec method to accurately encode RNA sequence information. In conjunction with a novel, word2vec-based encoding method, we use a graph neural network classifier that can accurately predict miRNA-mRNA interactions based on graph representation learning. As part of a comparative study, we evaluate three different node embedding approaches within the GraphTar framework and compare them with other state-of-the-art target prediction methods. The results show that the proposed method achieves similar performance to the best methods in the field and outperforms them on one of the datasets. Conclusions: In this study, a novel miRNA target prediction approach called GraphTar is introduced. Results show that GraphTar is as effective as existing methods and even outperforms them in some cases, opening new avenues for further research. However, the expansion of available datasets is critical for advancing the field towards real-world applications.
... We hypothesized that the reduction of p53 mRNA level was due to miRNAs targeting p53 expression. According to miRTarBase (http://mirtarbase.mbc.nctu.edu.tw/php/index.php) and the literature (Ishiguro H et al. 2014;Rokavec M et al. 2014; we analyzed the expression of potential miRNAs targeting p53 (miR-25, miR-30d, miR-125a, and miR-222) and a miRNA targeted by p53 (miR-22) (Tsuchiya N et al. 2011;Lin J et al. 2014). To quantify miRNA expressions by the method 2 (-ΔΔCt) we tested the expression of reference miRNA (RNU6, SNORD65, and SNORD95) (Supplementary figure 6). ...
Thesis
Due to the efficiency of treatments, the 5-year overall survival rate for patients with gastric cancer (GC) is approximately 15%. Currently, there is no stratification of patients to prescribe an effective treatment protocol.During my thesis, I established the role of HDAC4 in the sensitivity of GC cells to Cisplatin. I have shown that this response seems to depend on the type of GC (intestinal or diffuse) and the p53 status of cancer cells. I emphasized the interest of combining an HDAC inhibitor (SAHA) with platinum derivative chemotherapies (PDC: Cisplatin, Oxaliplatin) to promote their cytotoxic effects. Interestingly, I observed that the response to combination treatments is different depending on the p53 status of the cancer cells.These results open new perspectives in the use of PDC + SAHA combination therapies in GC. The p53 factor that is often mutated in GC could be a therapeutic marker for a such treatment protocol.
... In this situation, the TAS suppressed tumor growth [42]. miR-145 is regulated by TP53 in various cancers including pancreatic cancer [43]. miR-145 targets multiple mRNAs encoding proteins important in tumor progression. ...
Article
Full-text available
Approximately 90% of pancreatic cancers are pancreatic ductal adenocarcinomas (PDAC). PDAC is the fourth leading cause of cancer death world-wide. Therapies for PDAC are largely ineffective due to the dense desmoplastic tumor microenvironment which prevents chemotherapeutic drugs and small molecule inhibitors from exerting effective anti-cancer effects. In this review, we will discuss the roles of TP53 and miRs on the PDAC tumor microenvironment and how loss of the normal functions of TP53 promote tumor progression. The TP53 gene is mutated in approximately 50% of pancreatic cancers. Often, these TP53 mutations are point mutations which confer additional functions for the TP53 proteins. These are called gain of function (GOF) mutations (mut). Another class of TP53 mutations are deletions which result in loss of the TP53 protein; these are referred to TP53-null mutations. We have organized this review into various components/properties of the PDAC microenvironment and how they may be altered in the presence of mutant TP53 and loss of certain miR expression.
... Nevertheless, all miRNAs were found to intervene in signalling pathways usually deregulated in CRC such as the ones of WNT, TGFβ, TP53, PI3K-AKT, Ras and MAPK. Various studies have already demonstrated the key role of miRNAs in regulating these pathways [4,20,67,69,[91][92][93][94][95] thus reinforcing our findings. Furthermore, Reid and colleagues showed that many miRNAs deregulated in CRC were computationally mapped to targets involved in pathways related to tumour progression [96]. ...
... On the other hand, TP53INP1 induces p53 protein phosphorylation at Ser-46 and activate p53, which is manifested as a positive feedback regulation of p53 [44]. Also, studies have also confirmed that p53 induces expression and/or maturation of several miRNAs, which leads to the repression of critical effector proteins [55,56]. Although our study identified the role of miR-106a and TP53INP1 in tumour metastasis, the complex interplay between p53-miRNA-TP53INP1 1 only begin to be explored further. ...
Article
Full-text available
Bone metastasis is one of the most serious complications in lung cancer patients. MicroRNAs (miRNAs) play important roles in tumour development, progression and metastasis. A previous study showed that miR-106a is highly expressed in the tissues of lung adenocarcinoma with bone metastasis, but its mechanism remains unclear. In this study, we showed that miR-106a expression is dramatically increased in lung cancer patients with bone metastasis (BM) by immunohistochemical analysis. MiR-106a promoted A549 and SPC-A1 cell proliferation, migration and invasion in vitro. The results of bioluminescence imaging (BLI), micro-CT and X-ray demonstrated that miR-106a promoted bone metastasis of lung adenocarcinoma in vivo. Mechanistic investigations revealed that miR-106a upregulation promoted metastasis by targeting tumour protein 53-induced nuclear protein 1 (TP53INP1)-mediated metastatic progression, including cell migration, autophagy-dependent death and epithelial–mesenchymal transition (EMT). Notably, autophagy partially attenuated the effects of miR-106a on promoting bone metastasis in lung adenocarcinoma. These findings demonstrated that restoring the expression of TP53INP1 by silencing miR-106a may be a novel therapeutic strategy for bone metastatic in lung adenocarcinoma.
Article
Full-text available
Background MicroRNAs (miRNAs) are short, non-coding RNA molecules that regulate gene expression by binding to specific mRNAs, inhibiting their translation. They play a critical role in regulating various biological processes and are implicated in many diseases, including cardiovascular, oncological, gastrointestinal diseases, and viral infections. Computational methods that can identify potential miRNA–mRNA interactions from raw data use one-dimensional miRNA–mRNA duplex representations and simple sequence encoding techniques, which may limit their performance. Results We have developed GraphTar, a new target prediction method that uses a novel graph-based representation to reflect the spatial structure of the miRNA–mRNA duplex. Unlike existing approaches, we use the word2vec method to accurately encode RNA sequence information. In conjunction with the novel encoding method, we use a graph neural network classifier that can accurately predict miRNA–mRNA interactions based on graph representation learning. As part of a comparative study, we evaluate three different node embedding approaches within the GraphTar framework and compare them with other state-of-the-art target prediction methods. The results show that the proposed method achieves similar performance to the best methods in the field and outperforms them on one of the datasets. Conclusions In this study, a novel miRNA target prediction approach called GraphTar is introduced. Results show that GraphTar is as effective as existing methods and even outperforms them in some cases, opening new avenues for further research. However, the expansion of available datasets is critical for advancing the field towards real-world applications.
Chapter
Colorectal cancer is the most common cancer worldwide and a major health problem due to its high mortality. The prognosis of patients depends primarily on the extent of the disease. Thus, early detection is the key prerequisite for improved survival. Unfortunately, the current screening programs are insufficient for global application due to the low sensitivity and specificity of fecal occult blood tests and the high costs and invasiveness of colonoscopies. Therefore, the development of a simple noninvasive and accurate test is highly desirable. In addition, novel biomarkers for the improved prediction of patients' prognosis and treatment response are necessary to improve the clinical stratification and therapy individualization. Today, small noncoding RNAs called microRNAs are well-described molecules deregulated in many cancer types and involved in numerous biological processes such as cell differentiation, proliferation, apoptosis, migration, angiogenesis, and cell cycle regulation. Importantly, they are stably present in different types of body fluids including blood serum, plasma, urine, or saliva, indicating that they could serve as valuable noninvasive biomarkers for early diagnosis and follow-up investigations. This chapter is a comprehensive examination of microRNAs’ involvement in the main signaling pathways and biological processes associated with colorectal cancer pathogenesis. Further, the most significant studies analyzing the diagnostic, prognostic, and predictive potential of tissue, stool, and circulating microRNAs are summarized and discussed.
Article
Full-text available
MDM4, also called MDMX or HDMX in humans, is an important negative regulator of the p53 tumor suppressor. MDM4 is overexpressed in about 17% of all cancers and more frequently in some types, such as colon cancer or retinoblastoma. MDM4 is known to be post-translationally regulated by MDM2-mediated ubiquitination to decrease its protein levels in response to genotoxic stress, resulting in accumulation and activation of p53. At the transcriptional level, MDM4 gene regulation has been less clearly understood. We have reported that DNA damage triggers loss of MDM4 mRNA and a concurrent increase in p53 activity. These experiments attempt to determine a mechanism for down-regulation of MDM4 mRNA. Here we report that MDM4 mRNA is a target of hsa-mir-34a (miR-34a). MDM4 mRNA contains a lengthy 3' untranslated region; however, we find that it is a miR-34a site within the open reading frame (ORF) of exon 11 that is responsible for the repression. Overexpression of miR-34a, but not a mutant miR-34a, is sufficient to decrease MDM4 mRNA levels to an extent identical to those of known miR-34a target genes. Likewise, MDM4 protein levels are decreased by miR-34a overexpression. Inhibition of endogenous miR-34a increased expression of miR-34a target genes and MDM4. A portion of MDM4 exon 11 containing this 8mer-A1 miR-34a site fused to a luciferase reporter gene is sufficient to confer responsiveness, being inhibited by additional expression of exogenous mir-34a and activated by inhibition of miR-34a. These data establish a mechanism for the observed DNA damage-induced negative regulation of MDM4 and potentially provide a novel means to manipulate MDM4 expression without introducing DNA damage.
Article
Full-text available
Significance Small RNAs can potently and precisely regulate gene expression; as a result, they have tremendous clinical potential. However, effective delivery of small RNAs to solid tumors has remained challenging. Here we report that a lipid/polymer nanoparticle can deliver small RNAs to treat autochthonous tumors in the so-called “KP” mouse model of lung cancer. Nanoparticles formulated with mimics of the p53-regulated miRNA miR-34a downregulated target genes and delayed tumor progression, while nanoparticles formulated with siRNA targeting Kirsten rat sarcoma viral oncogene homolog (siKras) slowed tumor growth and increased apoptosis. Notably, concurrent delivery of miR-34a and siKras increased anti-tumor effects, and led to tumor regression. These results demonstrate that small RNA therapies can impact solid lung tumor growth, and that targeted RNA combination therapies may be used to improve therapeutic response.
Article
Full-text available
Background: Recent studies have demonstrated that microRNAs are stably detectable in plasma/serum because of their binding to specific proteins or being packaged in secretory particles. This study was designed to detect novel microRNAs in plasma for cancer detection and monitoring using microRNA array-based approaches in oesophageal squamous cell carcinoma (ESCC) patients. Methods: Through the integration of two Toray 3D-Gene microRNA array-based approaches to compare plasma microRNA levels between ESCC patients and healthy volunteers and between preoperative and postoperative ESCC patients, we identified a novel plasma biomarker in ESCC. Results: (1) Eight upregulated and common microRNAs (miR-15b, 16, 17, 25, 19b, 20a, 20b, and 106a) were selected using two high-resolution microRNA array approaches. (2) Test-scale analyses by quantitative RT-PCR validated a significant higher levels of plasma miR-19b (P=0.0020) and miR-25 (P=0.0030) in ESCC patients than controls. However, a significant correlation was observed between plasma miR-19b levels and concentrations of red blood cells (P=0.0073) and haemoglobin (P=0.0072). (3) miR-25 expression was found to be significantly higher in ESCC tissues (P=0.0157) and ESCC cell lines (P=0.0093) than in normal tissues and fibroblasts. (4) In a large-scale validation analysis, plasma miR-25 levels were significantly higher in 105 preoperative (P<0.0001) ESCC patients who underwent curative oesophagectomy and 20 superficial ESCC patients who underwent endoscopic resection (P<0.0001) than in 50 healthy volunteers. (5) Plasma miR-25 levels were significantly reduced in postoperative samples than in preoperative samples (P<0.0005) and were significantly increased during ESCC recurrences (P=0.0145). Conclusions: Plasma miR-25 might be a clinically useful biomarker for cancer detection and the monitoring of tumour dynamics in ESCC patients.
Article
Full-text available
The tumor suppressor p53 is one of the most frequently mutated genes in human cancers. MicroRNAs (miRNAs) are small non-protein coding RNAs that regulate gene expression on the post-transcriptional level. Recently, it was shown that p53 regulates the expression of several miRNAs, thereby representing an important mechanism of p53 signaling. Several independent studies identified the members of the miR-34 family as the most prevalent p53-induced miRNAs. miR-34s are frequently silenced in variety of tumor entities, suggesting that they are important tumor suppressors. Indeed, ectopic expression of miR-34s inhibits proliferation, epithelial to mesenchymal transition, migration, invasion, and metastasis of various cancer cell entities. Moreover, delivery or re-expression of miR-34 leads to notable repression of tumor growth and metastasis in cancer mouse models, and may therefore represent an efficient strategy for future cancer therapeutics. Besides their crucial functions in cancer, members of the miR-34 family also play important roles in spermatogenesis, stem cell differentiation, neuronal development, aging, and cardiovascular functions. Consequently, miR-34 has also been implicated in various non-cancerous diseases, such as brain disorders, osteoporosis, and cardiovascular complications.
Article
Through a PCR‐based differential screening method, cyclin G was identified as a novel transcriptional target of the p53 tumor suppressor gene product. In both a mouse p53 temperature‐sensitive leukemic cell line and mouse embryonic fibroblasts (MEF) after gamma‐irradiation, cyclin G mRNA was rapidly induced. MEF from a p53‐deficient mouse expressed cyclin G at a level > 10‐fold lower than that from a wild‐type mouse. Using a DNA binding assay, a specific p53 binding site was identified upstream from the cyclin G gene, which functioned as a p53‐dependent cis‐acting element in a transient transfection assay. These results suggest that cyclin G might participate in a p53‐mediated pathway to prevent tumorigenesis.
Article
p53 suppresses tumor progression and metastasis. Epithelial-mesenchymal transition (EMT) is a key process in tumor progression and metastasis. The transcription factors ZEB1 and ZEB2 promote EMT. Here we show that p53 suppresses EMT by repressing expression of ZEB1 and ZEB2. By profiling 92 primary hepatocellular carcinomas (HCCs) and 9 HCC cell lines, we found that p53 upregulates microRNAs including miR-200 and miR-192 family members. The miR-200 family members transactivated by p53 then repress ZEB1/2 expression. p53-regulated miR-192 family members also repress ZEB2 expression. Inhibition or over-expression of the microRNAs affects p53-regulated EMT by altering ZEB1 and ZEB2 expression. Our findings indicate that p53 can regulate EMT, and that p53-regulated microRNAs are critical mediators of p53-regulated EMT. Citation Format: Taewan Kim, Tae Jin Lee, Young-Jun Jeon, Pascal Pineau, Anne Dejean, Carlo Maria Croce. p53 regulates epithelial-mesenchymal transition through microRNAs targeting ZEB1 and ZEB2 [abstract]. In: Proceedings of the AACR Special Conference on Noncoding RNAs and Cancer; 2012 Jan 8-11; Miami Beach, FL. Philadelphia (PA): AACR; Cancer Res 2012;72(2 Suppl):Abstract nr A18.