ArticlePDF Available

Oncofetal H19-derived miR-675 regulates tumor suppressor RB in human colorectal cancer

Authors:

Abstract and Figures

H19 is an imprinted oncofetal non-coding RNA recently shown to be the precursor of miR-675. The pathophysiological roles of H19 and its mature product miR-675 to carcinogenesis have, however, not been defined. By quantitative reverse transcription-polymerase chain reaction, both H19 and miR-675 were found to be upregulated in human colon cancer cell lines and primary human colorectal cancer (CRC) tissues compared with adjacent noncancerous tissues. Subsequently, the tumor suppressor retinoblastoma (RB) was confirmed to be a direct target of miR-675 as the microRNA suppressed the activity of the luciferase reporter carrying the 3′-untranslated region of RB messenger RNA that contains the miR-675-binding site. Suppression of miR-675 by transfection with anti-miR-675 increased RB expression and at the same time, decreased cell growth and soft agar colony formation in human colon cancer cells. Reciprocally, enhanced miR-675 expression by transfection with miR-675 precursor decreased RB expression, increased tumor cell growth and soft agar colony formation. Moreover, the inverse relationship between the expressions of RB and H19/miR-675 was also revealed in human CRC tissues and colon cancer cell lines. Our findings demonstrate that H19-derived miR-675, through downregulation of its target RB, regulates the CRC development and thus may serve as a potential target for CRC therapy. © The Author 2009. Published by Oxford University Press. All rights reserved. For Permissions, please email: [email protected] /* */
miR-675 directly targets on RB protein. ( A ) Predicted binding of miR-675 with the 3 # -UTR of RB mRNA. ( B ) The firefly luciferase activity in human colon cancer cells after cotransfection with pMIR-Rb/pMIR-RB-mut 3 # -UTR reporter construct and anti-miR-675 (Clone A, MIP101 and HT-29) or miR-675 precursor (SW480). The luciferase activity was measured by dual-luciferase reporter assay (Promega) and was normalized to Renilla luciferase activity. Mean ± SEM, n 5 3, ÃÃ P , 0.01. ( C ) The effect of miR-675 transfection on the expression of RB protein in human colon cancer cells. The cells were transfected with anti-miR-675 or miR-675 precursor for 24 h. Thereafter, the cells were lysed for western blot analysis of the RB protein level. Experiments were repeated at least three times with similar results and only one of the representative results was shown. b -Actin was used as the loading control. The number beneath the protein band is the relative expression of RB protein. The intensity of the bands was quantitated by densitometry. The expression of RB protein was first normalized with the expression of b -actin in the sample. Relative expression of RB protein in cells with miR-675 precursor or anti-miR-675 transfection was then calculated in relation to that of the respective control, of which the expression is designated as 1.0. Anti-control: anti-miRNA control; Pre-control: miRNA precursor control. ( D ) The RB protein level in human colon cancer cells as assessed by western blotting. Experiments were repeated three times with results similar to the one shown. b -Actin was used as a loading control, Mean ± SEM.
… 
Content may be subject to copyright.
Carcinogenesis vol.31 no.3 pp.350–358, 2010
doi:10.1093/carcin/bgp181
Advance Access publication November 19, 2009
Oncofetal H19-derived miR-675 regulates tumor suppressor RB in human colorectal
cancer
Wing Pui Tsang, Enders K.O.Ng
1
, Simon S.M.Ng
2
,
Hongchuan Jin
1
, Jun Yu
1
, Joseph J.Y.Sung
1
and Tim Tak
Kwok
Department of Biochemistry,
1
Institute of Digestive Disease, Li Ka Shing
Institute of Health Sciences, State Key Laboratory in Oncology in South China
and
2
Department of Surgery, Chinese University of Hong Kong, Shatin, N.T.,
Hong Kong Special Administrative Region, China
To whom correspondence should be addressed. Science Centre, Department
of Biochemistry, Chinese University of Hong Kong, Hong Kong Special
Administrative Region, China. Tel: þ852 26036311; Fax: þ852 26037246;
Email: kwok2020@cuhk.edu.hk
H19 is an imprinted oncofetal non-coding RNA recently shown to
be the precursor of miR-675. The pathophysiological roles of H19
and its mature product miR-675 to carcinogenesis have, however,
not been defined. By quantitative reverse transcription–polymerase
chain reaction, both H19 and miR-675 were found to be upregu-
lated in human colon cancer cell lines and primary human
colorectal cancer (CRC) tissues compared with adjacent non-
cancerous tissues. Subsequently, the tumor suppressor retinoblas-
toma (RB) was confirmed to be a direct target of miR-675 as the
microRNA suppressed the activity of the luciferase reporter car-
rying the 3#-untranslated region of RB messenger RNA that con-
tains the miR-675-binding site. Suppression of miR-675 by
transfection with anti-miR-675 increased RB expression and at
the same time, decreased cell growth and soft agar colony forma-
tion in human colon cancer cells. Reciprocally, enhanced miR-675
expression by transfection with miR-675 precursor decreased RB
expression, increased tumor cell growth and soft agar colony for-
mation. Moreover, the inverse relationship between the expres-
sions of RB and H19/miR-675 was also revealed in human CRC
tissues and colon cancer cell lines. Our findings demonstrate that
H19-derived miR-675, through downregulation of its target RB,
regulates the CRC development and thus may serve as a potential
target for CRC therapy.
Introduction
Colorectal cancer (CRC) is the third most common cancer worldwide
with an estimated 1 million new cases and a half million deaths each
year (1). Screening for CRC from curable early stages has the poten-
tial to reduce both the incidence and mortality of the disease (2).
Although 5 year mortality rates of CRC have slightly declined over
the last three decades, there is still a pressing need to identify new
prognostic biomarkers and therapeutic targets for this disease. Fur-
thermore, the underlying pathophysiological mechanisms of CRC de-
velopment remain elusive (1–4).
MicroRNAs (miRNAs) are 19- to 25-nucleotide regulatory non-
coding RNAs that areinitially expressed as hairpin transcriptsof primary
miRNA. These primary miRNA hairpins are cleaved by two RNAase III
enzymes, Drosha and Dicer, to generate mature miRNAs. MiRNAs
regulate the expressions of a wide variety of genes by translation re-
pression or promoting RNA degradation and are important in the regu-
lation of various cellular processes, such as cellular proliferation,
differentiation and apoptosis (5–7). To date, .723 human miRNAs
are annotatedin the miRBase registry (miRBase version 11.0), but most
of the genes regulated by human miRNAs are not well defined.
Dysregulation of a specific spectrum of miRNAs in human malig-
nancies is frequently observed. Emerging evidence suggests miRNAs
function as both tumor suppressors and oncogenes. About 50% of
annotated human miRNAs located at chromosomal regions involved
in loss of heterozygosity, amplification or breakpoints that are asso-
ciated with cancers (5,7,8). The miRNAs downregulated in human
cancers indicate that they may function as tumor suppressors. Let-7,
which targets the oncogene RAS, has shown to be downregulated in
lung cancers (9). MiR-15 and 16, which target the antiapoptotic factor
BCL2, are downregulated in chronic lymphocytic leukemias (10).
Expression levels of miR-143 that targets ERK5 and miR-145 were
found to be decreased in colon cancer (11). In contrast, the miRNAs
upregulated in cancers may function as oncogenes. MiR-155 and its
host gene BIC are highly expressed in several types of B-cell lym-
phoma (12). The miR-17-92 cluster, which is located on chromosome
13q31, is activated by the oncogene c-Myc and is highly expressed in
B-cell lymphoma and lung cancer (13). Therefore, the importance of
miRNAs acting as a new layer of gene regulation in tumorigenesis is
emerging.
H19 is a paternally imprinted (maternally expressed) oncofetal
gene and is located on chromosome 11p15.5, close to the IGF II gene
locus. The H19 gene does not encode for a protein but instead codes
for a capped, spliced and polyadenylated 2.7 kb RNA (14–16). H19 is
highly expressed from the early stages of embryogenesis to fetal life
in many organs including the fetal adrenal, liver and placenta but is
nearly completely downregulated postnatally (17).
Emerging evidence showed that H19 expression was upregulated in
many cancers including CRC (18,19), hepatocellular carcinoma
(20), testicular cancer (21), choriocarcinoma (22), esophageal cancer
(18), ovarian cancer (23), breast cancer (24) andbladder cancer (25,26),
with or without the loss of imprinting. Patients with more H19-
positive bladder cancer cells are potentially at higher risk of recurrent
disease (27). In the tumor formed by the injection of choriocarcinoma
Jar and JEG-3 cells into the nude mice, the H19 RNA level is higher
than those cells before the injection (28). Similarly, the H19 RNA
level is greatly enhanced in tumor of human bladder carcinoma cells
formed in nude mice (25). The overexpression of H19 in cancer
tissues hints for its oncogenic function, but the exact underlying
mechanism is still not clear. Recently, H19 was reported to be the
primary miRNA precursor of miR-675 in both human and mice (29).
As both H19 and miRNAs are believed to be involved in tumorio-
genesis, this prompted us to speculate that the tumoriogenesis process
induced by H19 may be mediated through miR-675. Therefore, in this
study, we investigated the pathophysiological roles of H19 and miR-
675 in CRC carcinogenesis. Furthermore, using in silico prediction
and in vitro functional assays, we confirmed retinoblastoma (RB)
protein as a putative direct target of miR-675. This verification of
the oncogenic function of H19-miR-675-RB in CRC suggests that
this pathway may serve as the potential target for cancer therapy.
Materials and methods
Human cell lines
The human colon cancer cell lines, including 228, CaCO2, Clone A, HCT116,
HT-29, MIP101, SW480, and normal colon fibroblast cell lines, including
CCD-112CoN, CCD-18Co, were maintained routinely in Dulbecco’s modified
Eagle’s medium supplemented with 10% fetal bovine serum and 2 mM
L-glutamine (Invitrogen, Carlsbad, CA) and were grown at 37°C in a 10%
CO
2
atmosphere.
Patient samples
Primary CRC and their adjacent non-cancerous tissues were collected from
30 patients who underwent either endoscopy or surgical removal of tumors at
the Prince of Wales Hospital, Hong Kong. All patients provided written in-
formed consent for the use of their tissues. This project was approved by the
Abbreviations: CRC, colorectal cancer; mRNA, messenger RNA; miRNA,
microRNA; MTT, 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bro-
mide; PCR, polymerase chain reaction; RB, retinoblastoma; siRNA, small
interfering RNA; UTR, untranslated region.
ÓThe Author 2009. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oxfordjournals.org 350
by guest on February 19, 2013http://carcin.oxfordjournals.org/Downloaded from
Joint The Chinese University of Hong Kong-New Territories East Cluster
Clinical Research Ethics Committee, Hong Kong. The median age of the patients
is 74 (58–89 years old). All are adenocarcinoma at stage II–IV. All tissues had
been histologically confirmed. Tissue samples were collected and immediately
snap frozen in liquid nitrogen and stored at 80°C until further analysis.
RNA extraction
Total RNA containing miRNAs was extracted from the tissues or cells using
the Trizol reagent (Invitrogen) followed by miRNeasy mini column
(miRNeasy Mini Kit, QIAGEN, Hilden, Germany) enrichment of miRNA.
In brief, tissues samples were homogenized in Trizol reagent and chloroform
was then added according to the manufacturer’s recommendations. The mix-
ture was centrifuged at 12 000gfor 15 min at 4°C, and the aqueous layer was
transferred into new tubes. Then, 1.5 volume of 100% ethanol was added to the
aqueous layer. The mixture was then applied to an miRNeasy mini column
(QIAGEN) and processed according to the manufacturer’s recommendations.
Total RNA was eluted with RNase-free water and stored at 80°C. DNase
treatment was carried out to remove any contaminating DNA. RNA concen-
trations were determined by NanoDrop spectrophotometry.
Quantitative reverse transcription–polymerase chain reaction
For detection of H19 and RB messenger RNA (mRNA), 1.5 lg total RNA was
reverse transcribed by Moloney murine leukemia virus reverse transcriptase
with oligo-dT primer according to the manufacturer’s instructions (Promega
Corporation, Madison, WI). For miR-675 detection, 3 lg total RNA was used
in the reverse transcription reaction by using the QuantMir RT Kit (System
Biosciences, Mountain View, CA). Quantitative real-time polymerase chain re-
action (PCR) was performed by using SYBR-green PCR Master Mix in a Fast
Real-time PCR 7500 System (Applied Biosystems, Foster City, CA). The mature
miR-675 DNA sequence was used as the forward primer, and the 3#universal
primer provided from the QuantiMir RT Kit as the reverse primer. The human U6
RNA was amplified in parallel as an internal control. For mRNA detection, the
gene-specific primers were: H19 (forward: 5#-TACAACCACTGCACTACCTG-
3#;reverse:5#-TGGAATGCTTGAAGGCTGCT-3#); RB (forward: 5#-AAGGA-
GACAAGTTCGCATGT-3#;reverse:5#-GCCGGTAATTGTCGTAGTTT-3#)
(30). b-Actin was amplified in parallel as the internal control. PCR reactions
were pe rformed at 95°C for 10 min, followed by 40 cycles of 95°Cfor15sand
60°C for 1 min. DCt was calculated by subtracting the Ct of U6 or b-actin RNA
from the Ct of miR-675 or the mRNA of interest, respectively. DDCt was then
calculated by subtracting the DCt of the control from the DCt of the treatment
group. Fold change of miRNA or mRNA was calculated by the equation 2
DDCt
.
Ectopic expression and gene silencing of H19 in cells
For the enhanced expression study, the full-length H19 complementary DNAwas
subcloned into pcDNA3.1 expression vector (Invitrogen). For the knockdown of
H19 expression, two complementary oligonucleotides for small hairpin RNA
targeting 5#-CATCAAAGACACCATCGGA-3#sequences were chemically syn-
thesized. The annealed hairpin small interfering RNA (siRNA) was subcloned
into pSilencer 2.1-U6 neo vector (Ambion, Austin, TX). The negative control
hairpin siRNA with no sequence homology to human genes provided by the
Fig. 1. Increased miR-675 expression in human CRCs. (A) Relative expressions of H19 mRNA and miR-675 in CRC tumor tissues and the adjacent non-
cancerous tissues. Statistical difference was analyzed by Wilcoxon signed-rank test (P50.001 for H19; P50.019 for miR-675). Both H19 mRNA and miR-675
levels were measured by quantitative reverse transcription–PCR. (B) Relative expressions of H19 mRNA (left panel) and miR-675 (right panel) in human normal
colon cell lines (CCD-18Co, CCD-112CoN) and colon cancer cell lines, Mean ± SEM, n53. (C) Positive correlation of H19 and miR-675 expression in human
colon cancer cells, r50.8784, P50.00035.
H19/miR-675 regulates RB in colon cancer
351
by guest on February 19, 2013http://carcin.oxfordjournals.org/Downloaded from
manufacturer (Ambion) was used as the negative control. A total of 1.5 10
5
cells were seeded in 35 mm tissue culture dishes for 24 h, followed by trans-
fection with 2 lg of each respective plasmid with lipofectamine (Invitrogen) for
24 h. The cells were then subjected to RNA extraction or functional assays.
Transfection with miR-675 precursor or inhibitor
Enhanced or knockdown expressions of miR-675 were performed by trans-
fection with miR-675 precursor (Ambion) or anti-miR-675 (Ambion), respec-
tively. A total of 5 10
4
or 750 cells were plated in 35 mm culture dishes or
24/96-well plates, respectively, for 24 h and then transfected with 40 nM of
miR-675 precursor or inhibitor with lipofectamine 2000 (Invitrogen) for 24 h.
Commercially available precursor/inhibitor control (Ambion) was transfected
in parallel. The cells were then subjected to RNA/protein extraction or further
functional assays.
MiRNA target predictions
Computer-based RNA22 miRNA target detection program was used to predict
the miR-675 potential binding sites of the target mRNA (http://cbcsrv.watson
.ibm.com/rna22.html). The DNA sequence of the 3#-untranslated region
(UTR) region of RB mRNA was obtained from Genbank of the National Center
for Biotechnology Information webpage (http://www.ncbi.nlm.nih.gov/).
Luciferase activity assay
The part of RB 3#-UTR containing the 5#and 3#flanking sequences as well as
miR-675-binding sequence was amplified by a pair of primers (RB F: 5#-CT-
CTACTAGTCGTCAGTATGGTCTAACAC-3#,RBR:5#-CTCTAAGCTT-
GCTAATGCAGCTGTTTTAA-3#) and subcloned into pMIR-REPORT
vector (Ambion) immediately downstream of the luciferase gene to form the
pMIR-RB-3#-UTR construct. Restriction digestion sequences were shown in
bold letters. The pMIR-RB-3#-UTR-mut reporter construct with point muta-
tions in seed sequence was synthesized using the site-directed mutagenesis kit
(Stratagene, La Jolla, CA). A total of 5 10
4
cells were seeded in 24-well
plates for 24 h and then cotransfected with 800 ng of pMIR constructs with or
without 40 nM of miR-675 precursor/inhibitor for 24 h. Each sample was also
cotransfected with 0.05 lg of pRL-CMV plasmid-expressing Renilla luciferase
to monitor the transfection efficiency. At 24 h posttransfection, the activity of
firefly luciferase was measured by using the dual-luciferase reporter assay
system as described by the manufacturer (Promega). Relative luciferase activ-
ity was normalized with renilla luciferase activity.
MTT cell growth assay
A total of 750 cells were seeded in each well of a 96-well plate for 24 h. The
cells were then transfected with 40 nM of miR-675 precursor or inhibitor for 24
h and allowed to grow for 5 days. Thereafter, the cells were incubated in 50 ll
of 0.1 mg/ml solution of 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium
bromide (MTT) at 37°C for 3 h and then lysed in 150 ll of dimethyl sulfoxide
at room temperature for 30 min. The absorbance in each well was measured at
580 nm by a microplate reader.
Soft agar colony formation assay
Soft agar plates were prepared in six-well plates with a bottom layer of 0.6%
Noble agar in serum-free Dulbecco’s modified Eagle’s medium. The cells were
first seeded in 35 mm tissue culture dishes for 24 h and then transfected with
40 nM of miR-675 precursor or anti-miR-675. After trypsinization, 500 cells
mixed with 0.3% Noble agar in 10% fetal calf serum-supplemented Dulbecco’s
modified Eagle’s medium were seeded as the top agar layer onto the agar plates.
The cells were then incubated in a 37°C incubator for 3 weeks. The number of
colonies was counted after the colonies were stained with 0.05% crystal violet
for 1 h and washed extensively with 1phosphate-buffered saline.
Western blot analysis
Cells were lyzed in Lammeli’s lysis buffer containing 1% Triton X-100 and
scraped by a cell lifter. A 25 lg of protein was resolved in 12% sodium dodecyl
sulfate–polyacrylamide gel electrophoresis minigel and transferred onto
Immobilon-P membrane (Millipore, Billerica, MA). Membranes were probed with
primary antibody against RB (Santa Cruz Biotechnology, Santa Cruz, CA) at room
temperature for 2 h, washed extensively with 0.1% Tween-20 in phosphate-
buffered saline and incubated with secondary antibody conjugated with horse-
radish peroxidase at 1:10000 dilution. The signals were visualized with enhanced
chemiluminescence (Amersham Life Science, Buckinghamshire, UK).
Statistical analysis
The data are expressed as themean ± SEM from at least threeindependent experi-
ments. Thedifference between two groups inreal-time PCR, MTTassay,soft agar
colony formation assay and luciferase reporter assay was analyzed by two-tailed
Student’s t-test. The difference was significant for Pvalue of ,0.05. The ex-
pressions of H19 and miR-675 in CRC tissues and their matched adjacent non-
cancerous tissues were compared by Wilcoxon signed-rank test. The correlations
of miR-675 with H19 and RB mRNA expressions were examined by Pearson
correlation. The difference was considered significant for P-values of ,0.05.
Results
Confirmation of H19 as the precursor of miR-675 in human colon
cancer cells
H19 is reported to be the primary precursor of miR-675 by Cai and
Cullen, in which transfection with H19 complementary DNA contain-
ing the pri-miR-675 hairpin increased the expression of mature
miR-675 in human kidney 293T cells (29). To verify their findings,
we transfected human colon cancer cells, Clone A, HT-29, MIP101
and SW480 cells, with the H19-expressing vector and found that miR-
675 expression was greatly increased as determined by quantitative
PCR. On the other hand, deprivation of H19 expression by siRNA-
mediated knockdown remarkably reduced miR-675 expression in all
four cell lines (supplementary Figure 1 is available at Carcinogenesis
Fig. 2. The effect of miR-675 on cell proliferation in human colon cancer cells. The miR-675 expression in cells after transfection with (A) anti-miR-675 or (B)
miR-675 precursor was validated by quantitative reverse transcription–PCR. Relative miR-675 expression in cells was compared with those with the control
inhibitor or precursor, respectively. The cells were transfected with anti-miR-675 or miR-675 precursor for 24 h and allowed to grow for 5 days followed by the
MTT assay. Relative cell proliferation in cells with (A) anti-miR-675 or (B) miR-675 precursor transfection was compared with those with the control inhibitor or
precursor. Anti-control: anti-miRNA control; Pre-control: miRNA precursor control, Mean ± SEM, n53. P-values (two tailed) were calculated by Student’s
t-test, P,0.05; P,0.01.
W.P.Tsang et al.
352
by guest on February 19, 2013http://carcin.oxfordjournals.org/Downloaded from
Fig. 3. The effect of miR-675 on the clonogenicity in soft agar of human colon cancer cells. The cells were transfected with anti-miR-675/miR-675 precursor
for 24 h. After transfection, 500 cells were plated in 0.3% soft agar for 3 weeks. Anti-control: anti-miRNA control; Pre-control: miRNA precursor control,
Mean ± SEM, n53, P,0.05; P,0.01.
H19/miR-675 regulates RB in colon cancer
353
by guest on February 19, 2013http://carcin.oxfordjournals.org/Downloaded from
Online). Transfection with control expression vector or siRNA had no
effect on the miR-675 level in cells (data not shown). Thus, the results
were in agreement with the findings of by Cai et al. (29) that the
oncofetal H19 RNA is the primary precursor of miR-675.
Increased expression of H19 and miR-675 in primary CRC tissues and
in human colon cancer cell lines
We examined the expression of H19 and miR-675 in 20 pairs of
human CRC tissues and their adjacent normals. Both H19 and miR-
675 levels were significantly elevated in the CRC tissues than in the
adjacent non-cancerous tissues (P50.001 for H19 and P50.019 for
miR-675; Figure 1A). These results suggest that both H19 and miR-675
expressions were significantly overexpressed in human CRC. We then
examined the expressions of H19 and miR-675 in a panel of the fol-
lowing seven human colon cancer cell lines: HT-29, SW480, 228,
CaCO2, Clone A, HCT116 and MCP101. Our results showed that both
H19 and miR-675 were highly expressed in all these seven cell lines in
comparison with human colon CCD-112CoN and CCD-18Co
Fig. 4. miR-675 directly targets on RB protein. (A) Predicted binding of miR-675 with the 3#-UTR of RB mRNA. (B) The firefly luciferase activity in human
colon cancer cells after cotransfection with pMIR-Rb/pMIR-RB-mut 3#-UTR reporter construct and anti-miR-675 (Clone A, MIP101 and HT-29) or miR-675
precursor (SW480). The luciferase activity was measured by dual-luciferase reporter assay (Promega) and was normalized to Renilla luciferase activity. Mean ±
SEM, n53, P,0.01. (C) The effect of miR-675 transfection on the expression of RB protein in human colon cancer cells. The cells were transfected with
anti-miR-675 or miR-675 precursor for 24 h. Thereafter, the cells were lysed for western blot analysis of the RB protein level. Experiments were repeated at
least three times with similar results and only one of the representative results was shown. b-Actin was used as the loading control. The number beneath the protein
band is the relative expression of RB protein. The intensity of the bands was quantitated by densitometry. The expression of RB protein was first normalized with
the expression of b-actin in the sample. Relative expression of RB protein in cells with miR-675 precursor or anti-miR-675 transfection was then calculated in
relation to that of the respective control, of which the expression is designated as 1.0. Anti-control: anti-miRNA control; Pre-control: miRNA precursor control.
(D) The RB protein level in human colon cancer cells as assessed by western blotting. Experiments were repeated three times with results similarto the one shown.
b-Actin was used as a loading control, Mean ± SEM.
W.P.Tsang et al.
354
by guest on February 19, 2013http://carcin.oxfordjournals.org/Downloaded from
fibroblast cells (Figure 1B) or human normal colon tissues (data not
shown). Furthermore, the expressions H19 and miR-675 in the cells
were correlated with each other (r50.8784; Figure 1C).
Functional effect of H19/miR-675 on cell proliferation and cellular
transformation in vitro
Overexpression of H19/miR-675 in human colon cancer cell lines and
primary CRC implied that H19/miR-675 might play a role in colo-
rectal carcinogenesis. To test this, we first investigated the functional
effect of miR-675 on cell proliferation and soft agar colony formation
of the following four selected human colon cancer cell lines: Clone A,
HT-29, MIP101 and SW480 cells. As shown in Figure 2, both knock-
down and enforced expression of miR-675 were effective. Knock-
down of miR-675 by transfection with anti-miR-675 suppressed the
tumor cell growth of all four cell lines. Approximately 20–25% re-
duction in cell growth was observed at 5 days after transfection (all
P-values ,0.05; Figure 2A). Additionally, enforced miR-675 ex-
pression by transfection with miR-675 precursor also significantly
increased 25–50% of cell growth in all four cell lines (all P-values
,0.05; Figure 2B).
Interestingly, miR-675 expression affected not only cell growth but
also malignant transformation as featured by the anchorage-indepen-
dent growth of cells in soft agar. We showed that miR-675 knockdown
by anti-miR-675 significantly reduced the clonogenicity of Clone A
(40%), HT-29 (50%), MIP101 (67%) and SW480 (50%) cells
in soft agar (all P-values ,0.05; Figure 3). On the contrary, increase
in clonogenicity was observed in Clone A (40%), HT-29 (40%),
MIP101 (70%) and SW480 (270%) cells upon transfection with
miR-675 precursor (all P-values ,0.05; Figure 3). These results pro-
vide strong evidence that miR-675 plays a role in promoting malig-
nant transformation in cells.
To elucidate whether miR-675 was the mediator for the oncogenic
function of H19, Clone A and SW480 cells were cotransfected with
H19 complementary DNA and anti-miR-675. A 40–50% increase in
the clonogenicity in soft agar was observed for cells only with H19
transfection, but such increase was, however, significantly abrogated
by the cotransfection with anti-miR-675 (supplementary Figure 2 is
available at Carcinogenesis Online). Results suggested that H19 ex-
hibited the effect on cellular transformation in human colon cancer
cells via miR-675.
MiR-675 targets RB expression
MiRNAs mainly exert their functions by targeting the 3#-UTR of the
protein-coding genes to induce mRNA degradation and/or transla-
tional repression. Using RNA22 miRNA target detection program
and setting the maximum number of the seed nucleotides as six in-
stead of seven (31), the 3#-UTR of RB mRNA was aligned with the
sequence of mature miR-675 and RB was identified to be one of the
potential targets of miR-675. The sequence alignments for miR-675
and the 3#-UTR of RB mRNA are shown in Figure 4A; miR-675
targets the nt4111–4134 of RB mRNA (GenBank accession no.
M15400.1).
To confirm whether the predicted miR-675 target site in the 3#-
UTR of RB mRNA was responsible for its regulation, the 3#-UTR of
RB mRNA flanking the entire putative target sequence or 3#-UTR
with mutated target sequence was subcloned into the firefly luciferase
reporter vector (pMIR-REPORT). The construct was then cotrans-
fected with anti-miR-675/anti-miRNA control in Clone A, HT-29,
MIP101 cells or miR-675 precursor/miRNA precursor control in
SW480 cells (the cells with the least miR-675 expression). Our results
showed that the relative luciferase activity of the pMIR-RB-3#-UTR
construct with anti-miR-675 was significantly increased in Clone A,
HT-29 and MIP101 cells (all P-values ,0.05). On the other hand,
enhanced miR-675 expression significantly decreased the relative lu-
ciferase activity in SW480 cells (P-values ,0.05). The changes in
the luciferase activity of pMIR-RB-3#-UTR upon the transfection
with miR-675 inhibitor or precursor were, however, not observed if
the miR-675-binding sequence in the reporter was mutated (Figure 4B).
By examining the RB protein level following enforced or inhibition of
miR-675 expression, our results indicated that miR-675 expression
inhibition increased the RB protein level, whereas ectopic miR-675
expression consistently decreased the RB protein level in all four cell
lines (Figure 4C). Similar results were found for RB mRNA (data not
shown). Taken together, the data from the luciferase activity assay and
western blot analysis strongly support that RB protein is a direct target
of miR-675.
The interrelationship for the expressions of H19, miR-675 and RB
was further verified in human colon cancer cells. As shown in Figure
1C, the expression of H19 is positively correlated with the level of
miR-675 in the human colon cancer cell lines. As the target of miR-
675, the level of RB protein also appears to be negatively correlated
with the levels of both H19 and miR-675 in the human colon cancer
cells (Figure 4D). Similar correction was also observed for RB mRNA
(data not shown).
The H19/miR-675 regulates RB to promote cellular transformation
As RB is a well-known tumor suppressor and hereby confirmed to be
one of the target genes of miR-675, we further investigated whether
the effect of H19/miR-675 on cellular transformation is through RB.
As expected, knockdown of RB by specific siRNA suppressed the RB
Fig. 5. The effect of miR-675/RB on soft agar clonogenicity in human colon cancer cells. The cells were transfected with RB siRNA (RBi) with or without
the anti-miR-675 for 24 h and then plated in 0.3% soft agar for 3 weeks. The sequence of RB siRNA duplex was adapted from Semizarov et al. (32). Mean ± SEM,
n53, P,0.05, P,0.01.
H19/miR-675 regulates RB in colon cancer
355
by guest on February 19, 2013http://carcin.oxfordjournals.org/Downloaded from
protein level (40% for Clone A and 55% for SW480 cells, data not
shown) and also increased the clonogenicity of cells in soft agar
(20% for Clone A and 200% for SW480; all P-values ,0.05).
Intriguingly, the decrease in clonogenicity by the anti-miR-675,
which is known to upregulate RB, was counteracted by the cotrans-
fection with RB siRNA (Figure 5). However, such changes were not
observed in the experiment using the control siRNA. This further
confirmed that the oncogenic role of H19/miR-675 is associated with
the downregulation of RB in the cancer cells.
Expression relationship of H19, miR-675 and RB in human primary
CRC tissues
The interrelationship for the expressions of H19, miR-675 and RB has
been defined in vitro. To verify their expression relationship in human
Fig. 6. The expressions of (A) H19 mRNA, (B) miR-675 and (C) RB mRNA in human CRC tissues. Relative expressions of H19 mRNA, miR-675 and RB
mRNA in the CRC tumor tissues (T) and the matched adjacent non-cancerous tissue (N) as detected by quantitative reverse transcription–PCR, Mean ± SEM,
n53. Asterisks represent CRC samples in which both H19 and miR-675 are downregulated. (D) Correlations of miR-675 with H19 (left panel) and Rb
mRNA (right panel) in CRC tissues.
W.P.Tsang et al.
356
by guest on February 19, 2013http://carcin.oxfordjournals.org/Downloaded from
CRC tissue samples, the expressions of H19, miR-675 and RB were
examined in an independent group of 10 pairs of human CRC tissues
with their matched adjacent non-cancerous tissues. Among this in-
dependent group analyzed, seven cases showed that the expressions of
both H19 and miR-675 were increased in the tumor tissues as com-
pared with their matched adjacent non-cancerous tissues, whereas the
RB mRNA level was also significantly reduced in the tumor tissues
(Figure 6). Such correlation is in agreement with the findings from the
present study showing that H19 is the precursor of miR-675 and the
miRNA downregulates RB expression in human colon cancer cells.
The results strongly support the probable implications of H19, miR-
675 and RB in CRC development even though other genetic factors
may also play significant role as not all CRC samples demonstrated
altered expressions of H19, miR-675 and RB.
Discussion
The present study is the first to establish the possible link between
H19/miR-675 and RB in CRC development. The overexpression of
H19 in CRC has previously been reported and it was suggested to be
due to the loss of imprinting of the gene (19). However, in this study,
we showed that both miR-675 and its precursor H19 were overex-
pressed in all seven colon cancer cell lines and the majority of primary
CRC tissues, but either was not or only very minimally expressed in
their adjacent non-cancerous tissues, suggesting the oncogenic role of
H19/miR675 in CRC development.
The oncogenic function of H19/miR-675 is featured by targeting
the well-known tumor suppressor RB. Molecular carcinogenesis of
colon cancer involves stepwise accumulation of epigenetic and ge-
netic alterations, including activating mutations of the K-Ras and
B-Raf oncogenes as well as inactivating mutations of APC and p53
tumor suppressor genes (3,33). The genetic pathways in colon carci-
nogenesis are complicated and some of the pathways may occur ran-
domly, concurrently with or even preferentially over the other
pathways (3,33,34). Recently, dysregulation of miRNAs was reported
in human CRC (11,34). The miRNAs may function as cross talk
between different colorectal carcinogenesis pathways and may there-
fore be the potential therapeutic targets. Such cross talk can be illus-
trated by the H19/miR-675/RB pathway identified in the present
study. The H19/miR-675/RB pathway was detected in human colon
cancer cell lines as well as in human CRC tissues. The role of this
pathway in CRC development is confirmed as: (i) ectopic expression
of H19-derived miR-675 promoted cell growth and induced malignant
transformation in human colon cancer cells and vice versa and (ii)
H19 and miR-675 were overexpressed in CRC tissue, whereas its
target gene, the tumor suppressor RB, was downregulated. Like
miR-675, miR-106a was also found to be overexpressed in colon
carcinoma that did not express RB (8). RB is functionally inactivated
in the majority of human cancers, subsequently leading to dysregula-
tion of cell cycles and aggressive tumor proliferation (35–38). The
role of RB in cell cycle regulation is mainly through its interaction
with transcription factor E2F. As E2F was shown to activate H19 (39),
it is therefore of interest in the future to see if there may be feedback
loop in the regulation of H19/miR-675/RB pathway.
The oncogenic role of H19 is associated with its function as the
precursor of miR-675. Even though H19 is known to have functions
related to cancer development, the underlying mechanism is still un-
clear. Although some of the H19 target genes have been identified, the
way that H19 may interact with these target genes is also still unclear.
It is believed that H19 RNA may interact directly with its target genes
or indirectly through some of the H19 interaction proteins (14–16,40).
In any case, a clear link between H19 and its target gene has yet to be
reported. The identification of H19 as the precursor of miR-675 and
the proof that the miRNA mediates the oncogenic function of H19
may provide a new perspective for the future investigation of the
action mechanism of H19. In fact, long-transcript non-coding RNA
to act as the precursor of miRNAs has been demonstrated previously,
e.g. lin 4 from lin-14 mRNA in Caenorhabditis elegans (41) and in
miR-155 from the transcript of proto-oncogene BIC (12). Neverthe-
less, the present study is the first to confirm the importance of the
miR-675 pathway in the biological function of H19.
In summary, the H19-derived miR-675 miRNA, by targeting tumor
suppressor RB, is proved to be oncogenic by promoting cell growth
and malignant transformation in human colon cancer cells. The upre-
gulation of H19 and miR-675 in CRC suggests that both H19 and
miR-675 are important factors in the tumorigenesis of CRC and that
they may also serve as potential prognosis markers as well as potential
targets for cancer therapy.
Supplementary material
Supplementary Figures 1 and 2 can be found at http://carcin
.oxfordjournals.org/
Funding
Hong Kong Research Grants Council (Earmarked Grant CUHK4270/
04M); Institute of Digestive Disease, Chinese University of Hong
Kong.
Acknowledgements
Conflict of Interest Statement: None declared.
References
1. Parkin,D.M. et al. (2005) Global cancer statistics, 2002. CA Cancer J.
Clin.,55, 74–108.
2. Walsh,J.M. et al. (2003) Colorectal cancer screening: scientific review.
JAMA,289, 1288–1296.
3. Fearnhead,N.S. et al. (2002) Genetics of colorectal cancer: hereditary as-
pects and overview of colorectal tumorigenesis. Br. Med. Bull.,64, 27–43.
4. Wolpin,B.M. et al. (2008) Systemic treatment of colorectal cancer. Gastro-
enterology,134, 1296–1310.
5. Garzon,R. et al. (2006) MicroRNA expression and function in cancer.
Trends Mol. Med.,12, 580–587.
6. Jovanovic,M. et al. (2006) miRNAs and apoptosis: RNAs to die for. On-
cogene,25, 6176–6187.
7. Zhang,B. et al. (2007) microRNAs as oncogenes and tumor suppressors.
Dev. Biol.,302, 1–12.
8. Volinia,S. et al. (2006) A microRNA expression signature of human solid
tumors defines cancer gene targets. Proc. Natl Acad. Sci. USA,103, 2257–
2261.
9. Johnson,S.M. et al. (2005) RAS is regulated by the let-7 microRNA family.
Cell,120, 635–647.
10. Cimmino,A. et al. (2005) miR-15 and miR-16 induce apoptosis by target-
ing BCL2. Proc. Natl Acad. Sci. USA,102, 13944–13949.
11. Michael,M.Z. et al. (2003) Reduced accumulation of specific microRNAs
in colorectal neoplasia. Mol. Cancer Res.,1, 882–891.
12. Eis,P.S. et al. (2005) Accumulation of miR-155 and BIC RNA in human B
cell lymphomas. Proc. Natl Acad. Sci. USA,102, 3627–3632.
13. He,L. et al. (2005) A microRNA polycistron as a potential human onco-
gene. Nature,435, 828–833.
14. Ayesh,S. et al. (2002) Possible physiological role of H19 RNA. Mol. Car-
cinog.,35, 63–74.
15. Matouk,I.J. et al. (2007) The H19 non-coding RNA is essential for human
tumor growth. PLoS ONE,2, e845.
16. Brannan,C.I. et al. (1990) The product of the H19 gene may function as an
RNA. Mol. Cell. Biol.,10, 28–36.
17. Lustig,O. et al. (1994) Expression of the imprinted gene H19 in the human
fetus. Mol. Reprod. Dev.,38, 239–246.
18. Hibi,K. et al. (1996) Loss of H19 imprinting in esophageal cancer. Cancer
Res.,56, 480–482.
19. Cui,H. et al. (2002) Loss of imprinting in colorectal cancer linked to hypo-
methylation of H19 and IGF2. Cancer Res.,62, 6442–6446.
20. Ariel,I. et al. (1998) Imprinted H19 oncofetal RNA is a candidate tumour
marker for hepatocellular carcinoma. Mol. Pathol.,51, 21–25.
21. Verkerk,A.J. et al. (1997) Unique expression patterns of H19 in human
testicular cancers of different etiology. Oncogene,14, 95–107.
22. Lustig-Yariv,O. et al. (1997) The expression of the imprinted genes H19
and IGF-2 in choriocarcinoma cell lines. Is H19 a tumor suppressor gene?
Oncogene,15, 169–177.
H19/miR-675 regulates RB in colon cancer
357
by guest on February 19, 2013http://carcin.oxfordjournals.org/Downloaded from
23. Tanos,V. et al. (1999) Expression of the imprinted H19 oncofetal RNA in
epithelial ovarian cancer. Eur. J. Obstet. Gynecol. Reprod. Biol.,85, 7–11.
24. Lottin,S. et al. (2002) Overexpression of an ectopic H19 gene enhances the
tumorigenic properties of breast cancer cells. Carcinogenesis,23, 1885–
1895.
25. Elkin,M. et al. (1995) The expression of the imprinted H19 and IGF-2
genes in human bladder carcinoma. FEBS Lett.,374, 57–61.
26. Byun,H.M. et al. (2007) Examination of IGF2 and H19 loss of imprinting in
bladder cancer. Cancer Res.,67, 10753–10758.
27. Ariel,I. et al. (2000) The imprinted H19 gene is a marker of early recur-
rence in human bladder carcinoma. Mol. Pathol.,53, 320–323.
28. Rachmilewitz,J. et al. (1995) H19 expression and tumorigenicity of cho-
riocarcinoma derived cell lines. Oncogene,11, 863–870.
29. Cai,X. et al. (2007) The imprinted H19 noncoding RNA is a primary micro-
RNA precursor. RNA,13, 313–316.
30. Perez,D.S. et al. (2008) Gene expression changes associated with altered
growth and differentiation in benzo[a]pyrene or arsenic exposed normal
human epidermal keratinocytes. J. Appl. Toxicol.,28, 491–508.
31. Kruger,J. et al. (2006) RNAhybrid: microRNA target prediction easy, fast
and flexible. Nucleic Acids Res.,34, W451–W454.
32. Semizarov,D. et al. (2004) siRNA-mediated gene silencing: a global ge-
nome view. Nucleic Acids Res.,32, 3836–3845.
33. Souglakos,J. (2007) Genetic alterations in sporadic and hereditary colorectal
cancer: implementations for screening and follow-up. Dig. Dis.,25,919.
34. Bandres,E. et al. (2006) Identification by real-time PCR of 13 mature
microRNAs differentially expressed in colorectal cancer and non-tumoral
tissues. Mol. Cancer.,5, 29.
35. Khidr,L. et al. (2006) RB, the conductor that orchestrates life, death and
differentiation. Oncogene,25, 5210–5219.
36. Scambia,G. et al. (2006) RB family members as predictive and prognostic
factors in human cancer. Oncogene,25, 5302–5308.
37. DeGregori,J. (2004) The rb network. J. Cell Sci.,117, 3411–3413.
38. Bosco,E.E. et al. (2007) RB in breast cancer: at the crossroads of tumor-
igenesis and treatment. Cell Cycle,6, 667–671.
39. Berteaux,N. et al. (2005) H19 mRNA-like noncoding RNA promotes breast
cancer cell proliferation through positive control by E2F1. J. Biol. Chem.,
280, 29625–29636.
40. Tsang,W.P. et al. (2007) Riboregulator H19 induction of MDR1-associated
drug resistance in human hepatocellular carcinoma cells. Oncogene,26,
4877–4881.
41. Wightman,B. et al. (1993) Posttranscriptional regulation of the hetero-
chronic gene lin-14 by lin-4 mediates temporal pattern formation in
C. elegans.Cell,75, 855–862.
Received May 12, 2009; revised July 8, 2009; accepted July 17, 2009
W.P.Tsang et al.
358
by guest on February 19, 2013http://carcin.oxfordjournals.org/Downloaded from
... 34 Additionally, the H19-derived miR-675 has been implicated in colorectal cancer through downregulation of its target Rb. 35 Furthermore, depletion of linc17500 impedes cell migration in TKE2 cells and leads to reduced levels of phosphorylated Akt. Activation of the phosphoinositide 3-kinase (PI3K)/Akt signaling pathway, characterized by elevated expression of p-Akt, holds crucial significance in cell migration and exerts notable effects on cutaneous wound healing processes, including regeneration 36 and remodeling. ...
Article
Full-text available
Purpose: Epigenetic mechanisms orchestrate a harmonious process of corneal epithelial wound healing (CEWH). However, the precise role of long non-coding RNAs (lncRNAs) as key epigenetic regulators in mediating CEWH remains elusive. Here, we aimed to elucidate the functional contribution of lncRNAs in regulating CEWH. Methods: We used a microarray to characterize lncRNA expression profiling during mouse CEWH. Subsequently, the aberrant lncRNAs and their cis-associated genes were subjected to comprehensive Gene Ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. Quantitative reverse transcription-polymerase chain reaction (RT-qPCR) and western blot analyses were performed to determine the expression profiles of key markers during CEWH. The in vivo effects of linc17500 on this process were investigated through targeted small interfering RNA (siRNA) injection. Post-siRNA treatment, corneal re-epithelialization was assessed, alongside the expression of cytokeratins 12 and 14 (Krt12 and Krt14) and Ki67. Effects of linc17500 on mouse corneal epithelial cell (TKE2) proliferation, cell cycle, and migration were assessed by multicellular tumor spheroids (MTS), 5-ethynyl-2'-deoxyuridine (EdU), flow cytometry, and scratch-wound assay, respectively. Results: Microarray analysis revealed dysregulation of numerous lncRNA candidates during CEWH. Bioinformatic analysis provided valuable annotations regarding the cis-associated genes of these lncRNAs. In vivo experiments demonstrated that knockdown of linc17500 resulted in delayed CEWH. Furthermore, the knockdown of linc17500 and its cis-associated gene, CDC28 protein kinase regulatory subunit 2 (Cks2), was found to impede TKE2 cell proliferation and migration. Notably, downregulation of linc17500 in TKE2 cells led to suppression of the activation status of Akt and Rb. Conclusions: This study sheds light on the significant involvement of lncRNAs in mediating CEWH and highlights the regulatory role of linc17500 on TKE2 cell behavior. Translational relevance: These findings provide valuable insights for future therapeutic research aimed at addressing corneal wound complications.
... [141] Intraperitoneal, lymphatic, hematogenic, and other pathways may spread CRC. Over the past decade, lncRNAs, particularly those that promote tumor development and progression, such as PVT1, [142] p21-associated ncRNA DNA damage-activated RNA (PANDAR), [143] HOTAIR, [144] a regulator of reprogramming (ROR), [145] UCA1, [146] and H19, [147] contribute to normal intestinal function and CRC formation. Using two amphiphilic copolymers, PEI-PDLL and DSPE-MPEG, Jia et al. developed a treatment for co-incorporated into polymeric hybrid nanoparticles (CSNP) in 2021 that consists of siCCAT1 and curcumin (Cur) (Figure 7). ...
Article
Full-text available
This review article spotlights the burgeoning potential of using nanotherapeutic strategies to target long non‐coding RNAs (lncRNAs) in cancer cells. This updated discourse underlines the prominent role of lncRNAs in instigating cancer, facilitating its progression, and metastasis, validating lncRNAs′ potential for being effective diagnostic biomarkers and therapeutic targets. The manuscript offers an in‐depth examination of different strategies presently employed to modulate lncRNA expression and function for therapeutic purposes. Among these strategies, Antisense Oligonucleotides (ASOs), RNA interference (RNAi) technologies, and the innovative clustered regularly interspaced short palindromic repeats (CRISPR)‐based gene editing tools garner noteworthy mention. A significant section of the review is dedicated to nanocarriers and their crucial role in drug delivery. These nanocarriers′ efficiency in targeting lncRNAs in varied types of cancers is elaborated upon, validating the importance of targeted therapy. The manuscript culminates by reaffirming the promising prospects of targeting lncRNAs to enhance the accuracy of cancer diagnosis and improve treatment efficacy. Consequently, new paths are opened to more research and innovation in employing nanotherapeutic approaches against lncRNAs in cancer cells. Thus, this comprehensive manuscript serves as a valuable resource that underscores the vital role of lncRNAs and the various nano‐strategies for targeting them in cancer treatment. Future research should also focus on unraveling the complex regulatory networks involving lncRNAs and identifying fundamental functional interactions to refine therapeutic strategies targeting lncRNAs in cancer.
... H19-derived miR-675 serves as a target for CRC therapy via the molecular H19/miR-675/RB1 pathway in CRC [98]. As aforementioned, oncofetal H19 is the primary miRNA template for miR-675 and both promote CRC tumorigenesis [40]. ...
Article
Full-text available
Colorectal cancer (CRC) has been recorded amongst the most common cancers in the world, with high morbidity and mortality rates, and relatively low survival rates. With risk factors such as chronic illness, age, and lifestyle associated with the development of CRC, the incidence of CRC is increasing each year. Thus, the discovery of novel biomarkers to improve the diagnosis and prognosis of CRC has become beneficial. Long non-coding RNAs (lncRNAs) have been emerging as potential players in several tumor types, one among them is the lncRNA H19. The paternally imprinted oncofetal gene is expressed in the embryo, downregulated at birth, and reappears in tumors. H19 aids in CRC cell growth, proliferation, invasion, and metastasis via various mechanisms of action, significantly through the lncRNA-microRNA (miRNA)-messenger RNA (mRNA)-competitive endogenous RNA (ceRNA) network, where H19 behaves as a miRNA sponge. The RNA transcript of H19 obtained from the first exon of the H19 gene, miRNA-675 also promotes CRC carcinogenesis. Overexpression of H19 in malignant tissues compared to adjacent non-malignant tissues marks H19 as an independent prognostic marker in CRC. Besides its prognostic value, H19 serves as a promising target for therapy in CRC treatment.
... Moreover, some lncRNAs function as a decoy/ sponge or precursor of miRNAs [107,[111][112][113]. Such as H19, the oncofetal lncRNA is the precursor RNA of miR-675 [114,115]. The association of H19 in tumorigenesis and invasion is attributable to the regulation of miR-675 [116,117]. ...
Chapter
Full-text available
In the last few years, the amount of genomic sequence data has grown exponentially. A large number of non-coding RNAs (ncRNAs) have been identified from bacteria to humans. ncRNAs are various and multi-faced; they can regulate gene expression through chromosomal, transcriptional, post-transcriptional, and translational levels and then participate in the whole process of development in different organisms. ncRNAs have been identified in the 1980s in Lepidoptera; they can play roles in growth, metamorphosis, metabolism, sex determination, reproduction, and immune response of insects. Now, the use of ncRNAs in pest control of Lepidoptera is also in process. This chapter will review the recent advance of ncRNAs in Lepidoptera and prospect the future studies of ncRNAs in insects.
... Furthermore, the abnormal expression of H19 is essential in the development of CRC (Yoshimizu et al., 2008). It has also been found that H19, apart from showcasing epigenetic regulation, also functions as a miRNA sponge and as a miRNA precursor in CRC (Tsang et al., 2010). ...
Chapter
Colorectal cancer (CRC) is among the most common cancers and is still the primary cause of cancer-related deaths worldwide. The metastases and proliferation of cancer cells depend on factors including the expression of genes and the upregulation of RNAs. Phospholipase A2 (PLA2) is an enzyme responsible for the release of lysophospholipids and active fatty acids from phospholipid pools in membranes, which supports the tumor microenvironment. The role of cyclooxygenase (COX) in the emergence and spread of tumors has recently received attention and it has been determined that COX-1 and COX-2 are two distinct isoforms of the enzyme COX, which is an enzyme with rate-limiting properties in the manufacture of prostaglandin (PG) from arachidonic acid (AA). COX-2 is an inducible enzyme that is persistently overexpressed and has an oncogenic influence in several malignancies, including CRC. Thus, this study explores the role of PLA2 in CRC, so that effective tests can be performed targeting COX and lipoxygenase (LOX) for cancer invasion. Additionally, this chapter also focuses on ncRNAs involved in regulatory roles in a wide range of malignant tumors, including CRC. Thus, this chapter summarizes the role of PLA2 and ncRNAs in CRC alongside exploring the possible correlation between ncRNAs and PLA2 in cancer progression.
Article
Full-text available
Cancer remains a major global health concern with high mortality rates mainly due to late diagnosis and poor prognosis. Long non-coding RNAs (lncRNAs) are emerging as key regulators of gene expression in human cancer, functioning through various mechanisms including as competing endogenous RNAs (ceRNAs) and indirectly regulating miRNA expression. LncRNAs have been found to have both oncogenic and tumor-suppressive roles in cancer, with the former promoting cancer cell proliferation, migration, invasion, and poor prognosis. Recent research has shown that lncRNAs are expressed in various immune cells and are involved in cancer cell immune escape and the modulation of the tumor microenvironment, thus highlighting their potential as targets for cancer immunotherapy. Targeting lncRNAs in cancer or immune cells could enhance the anti-tumor immune response and improve cancer immunotherapy outcomes. However, further research is required to fully understand the functional roles of lncRNAs in cancer and the immune system and their potential as targets for cancer immunotherapy. This review offers a comprehensive examination of the multifaceted roles of lncRNAs in human cancers, with a focus on their potential as targets for cancer immunotherapy. By exploring the intricate mechanisms underlying lncRNA-mediated regulation of cancer cell proliferation, invasion, and immune evasion, we provide insights into the diverse therapeutic applications of these molecules.
Article
Lung cancer (LC) imposes a significant burden, and is associated with high mortality and morbidity among malignant tumors. Aberrant expression of particular lncRNAs is closely linked to LC. LncRNA polymorphisms cause abnormal expression levels and/or structural dysfunction. They can affect the progression of cancer, survival, response to chemotherapy and recurrence rates in cancer patients. The present article provides a comprehensive overview of the effect of lncRNA genetic polymorphisms on LC. It is proposed that lncRNA-related variants can be used to predict cancer risk and therapeutic outcomes. More large-scale trials on diverse ethnic groups are required to validate the results, thus personalizing LC therapy based on lncRNA genotypes.
Article
Full-text available
Epidermal growth factor receptor (EGFR) is a cell surface protein that plays a vital role in regulating cell growth and division. However, certain tumors, such as colorectal cancer (CRC), can exhibit an overexpression of EGFR, resulting in uncontrolled cell growth and tumor progression. To address this issue, therapies targeting and inhibiting EGFR activity have been developed to suppress cancer growth. Nevertheless, resistance to these therapies poses a significant obstacle in cancer treatment. Recent research has focused on comprehending the underlying mechanisms contributing to anti-EGFR resistance and identifying new targets to overcome this striking challenge. Long noncoding RNAs (lncRNAs) are a class of RNA molecules that do not encode proteins but play pivotal roles in gene regulation and cellular processes. Emerging evidence suggests that lncRNAs may participate in modulating resistance to anti-EGFR therapies in CRC. Consequently, combining lncRNA targeting with the existing treatment modalities could potentially yield improved clinical outcomes. Illuminating the involvement of lncRNAs in anti-EGFR resistance mechanisms of cancer cells can provide valuable insights into the development of novel anti-EGFR therapies in several solid tumors.
Chapter
H19 is a long non-coding RNA (lncRNA) molecule that is encoded by the H19 gene located on chromosome 11 in humans and is exclusively expressed from the maternal allele. H19 is involved in the regulation of various cellular and biological processes, including embryonic development, cell growth, and differentiation. The H19 gene has been implicated in cancer development and progression, as it has been found to be dysregulated in a variety of cancers, including breast, liver, lung, and bladder cancer. Critically, H19 has been reported to act as both an oncogene and a tumor suppressor, indicating that its role in cancer is multifaceted and varies depending on the type of cancer and the specific cellular context. In addition to its biological functions, H19 is a key biomarker, potentially laying the groundwork for the implementation of treatment modalities in cancer as well as other age-related diseases. Despite significant progress in understanding the molecular mechanisms and functions of H19, much remains to be learned about its role in disease pathogenesis and potential therapeutic applications. This review provides an overview of the current knowledge about H19 lncRNA and its biological functions and discusses its potential as a therapeutic target for various cancers.KeywordsCancerH19Long non-coding RNA (lncRNA)
Article
Full-text available
Familial adenomatous polyposis and hereditary non-polyposis colorectal cancer are dominantly inherited conditions with 100% and 80% life-time risk of developing colorectal cancer, respectively. The genetic mutations responsible for these two conditions lie in the adenomatous polyposis coli (APC) and mismatch repair genes. These same genes also play a key role in the formation of sporadic colorectal cancers, which arise on a background of a similar spectrum of mutations to the hereditary cancers. This article examines the genetic mechanisms underlying the hereditary colorectal cancers, as well as genetic predisposition to colorectal cancer in the general population in the absence of a clear-cut genetic syndrome. Colorectal cancer arises as the cumulative effect of multiple mutations within the cell, allowing it to escape growth and regulatory control mechanisms. This step-wise progression of mutations facilitates the histological transition from normal mucosa to adenoma to carcinoma. The latter part of this paper focuses on the key genetic events underlying this process and provides an overview of the genetic mechanisms responsible for colorectal tumorigenesis. Colorectal cancer (CRC) is one of the commonest cancers in the Western world. While the majority of cases of CRC are sporadic, a significant minority occur as a result of an inherited genetic mutation. These inherited syndromes of CRC, together with the presence of a readily identifiable precursor lesion in the form of the adenomatous polyp, have greatly facilitated research into the genetic mechanisms responsible for colorectal tumorigenesis.
Article
Full-text available
Aims—To investigate the expression of the imprinted oncofetal H19 gene in human bladder carcinoma and to examine the possibility of using it as a tumour marker, similar to other oncofetal gene products. Methods—In situ hybridisation for H19 RNA was performed on 61 first biopsies of bladder carcinoma from Hadassah Medical Centre in Jerusalem. The intensity of the reaction and the number of tumour cells expressing H19 in each biopsy were evaluated in 56 patients, excluding biopsies with carcinoma in situ. The medical files were searched for demographic data and disease free survival. Results—More than 5% of cells expressed H19 in 47 of the 56 (84%) biopsies. There was a decrease in the number of cells expressing H19 with increasing tumour grade (loss of differentiation) (p = 0.03). Disease free survival from the first biopsy to first recurrence was significantly shorter in patients with tumours having a larger fraction of H19 expressing cells, controlling for tumour grade. This was also supported by the selective analysis of tumour recurrence in patients with grade I tumours. Conclusions—It might be possible to use H19 as a prognostic tumour marker for the early recurrence of bladder cancer. In addition, for the gene therapy of bladder carcinoma that is based on the transcriptional regulatory sequences of H19, the expression of H19 in an individual biopsy could be considered a predictive tumour marker for selecting those patients who would benefit from this form of treatment.
Article
The maternally expressed H19 gene is transcribed as an untranslated RNA that serves as a riboregulator. We have previously reported that this transcript accumulates in epithelial cells in ∼10% of breast cancers. To gain further insight on how the overexpression of the H19 gene affects the phenotype of human breast epithelial cells, we investigated the oncogenic potential of RNA that was abundantly expressed from MDA-MB-231 breast cancer cells stably transfected with the genomic sequence of the human H19 gene. The amount of H19 RNA did not affect cell proliferation capacity, timing of cell cycle phases or anchorage-dependent ability of H19-transfected clones in vitro. But in anchorage-independent growth assays the H19-recombined cells formed more and larger colonies in soft-agar versus control cells. To explore this phenotypic change, we analysed tumour development after subcutaneous injection of H19-recombined cells into scid mice. Results showed that H19 overexpression promotes tumour progression. These data support the hypothesis that an overload of H19 transcript is associated with cells exhibiting higher tumorigenic phenotypes and therefore we conclude that the H19 gene has oncogenic properties in breast epithelial cells.
Article
The mouse H19 gene was identified as an abundant hepatic fetal-specific mRNA under the transcriptional control of a trans-acting locus termed raf. The protein this gene encoded was not apparent from an analysis of its nucleotide sequence, since the mRNA contained multiple translation termination signals in all three reading frames. As a means of assessing which of the 35 small open reading frames might be important to the function of the gene, the human H19 gene was cloned and sequenced. Comparison of the two homologs revealed no conserved open reading frame. Cellular fractionation showed that H19 RNA is cytoplasmic but not associated with the translational machinery. Instead, it is located in a particle with a sedimentation coefficient of approximately 28S. Despite the fact that it is transcribed by RNA polymerase II and is spliced and polyadenylated, we suggest that the H19 RNA is not a classical mRNA. Instead, the product of this unusual gene may be an RNA molecule.
Article
Certain embryonal tumors demonstrate a loss of heterozygosity at the parentally imprinted region of chromosome 11p15.5. It has been hypothesized that this implicates a tumor suppressor gene at this locus. The human H19 gene maps to 11p15.5, is expressed in fetal tissues including the placenta and is paternally imprinted. Here we show that the abundance of H19 transcripts in cells of two choriocarcinoma derived cell lines (JAr and JEG-3) differs greatly. While JAr cells express high levels of H19 RNA, the expression of H19 in JEG-3 cells is much lower than that of normal trophoblasts. Cells of these two cell lines were subcutaneously injected into nude mice with subsequent tumor formation. A fivefold increase in the H19 RNA level was measured in tumors derived from JEG-3 cell lines as compared to these cells before injection. However this increase in H19 RNA did not alter the clonogenicity in soft agar nor the growth rate of the cells derived from these tumors as compared to the original JEG-3 cells. Nevertheless, the cells retaining the elevated level of H19 transcripts were more tumorigenic than the original cells. We propose that there is a selection of cells expressing high levels of H19 from the total JEG-3 cell population during the microevolution of tumor formation. These observations, together with our previous publications on H19 expression in human cancers, do not support the notion of a tumor suppressor role for the H19 gene.
Article
The imprinted H19 gene is highly expressed in human embryos, fetal tissues and is nearly completely shut off in adults. However, it is reexpressed in a number of tumors including bladder carcinoma, demonstrating that H19 RNA is an oncofetal RNA. Tumors induced by injection of bladder carcinoma cell lines express H19 in contrast to the cells before injection. These observations support the notion of a positive correlation between H19 expression and bladder carcinoma. Loss of imprinting of H19 and IGF-2 was observed in samples of human bladder carcinoma.
Article
The H19 gene is a parenterally imprinted maternally expressed gene which has a pivotal role in embryogenesis and fetal development. It is tightly linked to the IGF-II gene on chromosome 11p15.5 which is reciprocally imprinted. We studied the expression of the human H19 by in situ hybridization in an embryo 35 days post coitus (dpc) and in a fetus from the second trimester of pregnancy. The expression pattern of H19 in the human fetal tissues was similar to its expression in the mouse, and paralleled, with some exceptions, the expression of IGF-II in human fetuses. Abundant expression was found in organs comprising the fetoplacental unit: the placenta, the fetal adrenal, and liver. The expression in the fetal adrenal cortex was most prominent in the definitive cortex and somewhat weaker in the fetal zone. Considerable expression of H19 was found in the fetal liver as early as 35 dpc and in the second trimester. Hematopoietic cells in fetal liver did not express the gene. Moderate expression of H19 was detected in the epithelium of the small intestines, in endometrial stroma and Fallopian tube. In the kidney conspicuous labeling of the metanephric blastema was noted, which was markedly reduced with differentiation to tubules. This pattern of expression is identical to that of IGF-II in the fetal kidney and is relevant to the evolution of Wilms' tumor. No expression of H19 was found in the neural tube of the first trimester embryo or in the developing fetal brain in the second trimester, nor were transcripts detected in the choroid plexus.(ABSTRACT TRUNCATED AT 250 WORDS)