ArticlePDF AvailableLiterature Review

Abstract and Figures

Ovarian cancer (OC) is the second most common and the most fatal gynecologic cancer in the United States. Over the last decade, various targeted therapeutics have been introduced but there has been no corresponding improvement in patient survival mainly because of the lack of effective early detection methods. microRNAs (miRs) are small, non-coding RNAs that regulate gene expression post-transcriptionally. Accumulating data suggest central regulatory roles of miRs in modulating OC initiation, progression, and metastasis. More recently, aberrant miR expression has been also associated with cancer stem cell (CSC) phenotypes and development of CSC chemo-resistance. Here, we review recent advances on miRs and OC metastasis and discuss the concept that miRs are involved in both CSC transformation and subsequent OC metastasis. Finally, we describe the prevalence of circulating miRs and assess their potential utilities as biomarkers for OC diagnosis, prognosis, and therapeutics.
Content may be subject to copyright.
MINI REVIEW ARTICLE
published: 10 June 2014
doi: 10.3389/fonc.2014.00143
Regulation of metastasis by microRNAs in ovarian cancer
Yongchao Wang 1, Sangmi Kim2and Il-man Kim1,3 *
1Vascular Biology Center, Medical College of Georgia, Georgia Regents University, Augusta, GA, USA
2Cancer Center, Medical College of Georgia, Georgia Regents University, Augusta, GA, USA
3Department of Biochemistry and Molecular Biology, Medical College of Georgia, Georgia Regents University, Augusta, GA, USA
Edited by:
Shailender S. Kanwar, University of
Michigan, USA
Reviewed by:
Santos Mañes, Consejo Superior de
Investigaciones Científicas, Spain
Vinesh KumarThidil Puliyappadamba,
University of Alabama, USA
*Correspondence:
Il-man Kim, Department of
Biochemistry and Molecular Biology,
Vascular Biology Center, Medical
College of Georgia, Georgia Regents
University, CB-3717, 1459 Laney
Walker Blvd, Augusta, GA 30912, USA
e-mail: ilkim@gru.edu
Ovarian cancer (OC) is the second most common and the most fatal gynecologic cancer
in the United States. Over the last decade, various targeted therapeutics have been intro-
duced but there has been no corresponding improvement in patient survival mainly because
of the lack of effective early detection methods. microRNAs (miRs) are small, non-coding
RNAs that regulate gene expression post-transcriptionally. Accumulating data suggest cen-
tral regulatory roles of miRs in modulating OC initiation, progression, and metastasis. More
recently, aberrant miR expression has been also associated with cancer stem cell (CSC)
phenotypes and development of CSC chemo-resistance. Here, we review recent advances
on miRs and OC metastasis and discuss the concept that miRs are involved in both CSC
transformation and subsequent OC metastasis. Finally, we describe the prevalence of cir-
culating miRs and assess their potential utilities as biomarkers for OC diagnosis, prognosis,
and therapeutics.
Keywords: ovarian cancer, miRs, cancer stem cells, epithelial–mesenchymal transition, extracellular matrix,
angiogenesis
INTRODUCTION
Ovarian cancer (OC) is the second most common gynecologic
cancer and the deadliest malignancy among women in the United
States (1). The current standard treatment rests on surgery fol-
lowed by platinum-based chemotherapy. Early disease may be
successfully removed with surgery alone, but most patients with
OC are diagnosed at later stages. Advanced diseases, especially
those metastasized, require complex treatment and management,
accounting for high morbidity and mortality associated with OC.
Cancer antigen 125 (CA-125), the currently available OC diag-
nostic marker, can only be adopted in advanced stages and has
limited utility due to its lack of sensitivity and specificity (26).
Therefore, development of novel biomarkers for early detection of
OC is imperative and, toward the goal, research efforts to under-
stand the detailed mechanism underlying its tumorigenesis and
metastasis are warranted.
microRNAs (miRNAs or miRs) are small non-coding RNAs.
They are first transcribed as primary miR transcripts (pri-miRs)
and cropped by the Drosha complex into premature miRs (pre-
miRs). Following the cleavage, exportin-5 mediates the nuclear
translocation of the pre-miRs, which are further processed by the
Dicer complex into mature miRs. The mature miRs are incor-
porated into RNA-induced silencing complex to execute gene-
silencing effect either by repressing the translation or directly
inhibiting messenger RNA (mRNA) stability. Notably, the 30
untranslated regions (UTRs) of each mRNA usually have mul-
tiple miR binding sites and conversely each miR has multiple or
even up to hundreds of target mRNAs as well. Therefore, even the
subtle change of miR expression levels could be amplified in effect
and consequently alters the functionality and phenotype of tar-
geted cells by adjusting their repertoires. The abnormal expression
of miRs in cancer leads to significant pathological consequences.
Oncogenesis is conferred either by overexpression of miRs tar-
geting tumor suppressors or loss of miRs that act as tumor sup-
pressors. Recently, the involvement of miRs in tumorigenesis and
metastasis of OC has been increasingly reported and potential
utility of miRs in early detection of OC has been proposed (715).
microRNAs AND OVARIAN CANCER STEM CELLS
A subpopulation of stem-like cells have been isolated and identi-
fied in several distinct malignancies. These stem-like cells, named
cancer stem cells (CSCs), are characterized as possessing the
unlimited self-renewal capacity and pluripotency and thus, they
could differentiate into various cell types and confer the hetero-
geneity of tumor cells (16). It is speculated that CSCs are evolved
from resident stem cells harbored in tissues and are suggested
to be genetically dynamic during tumor progression (17). Alter-
natively, epithelial–mesenchymal transition (EMT) could render
CSC formation as illustrated in Figures 1A,B.
Since Bapat et al. first reported the involvement of CSCs in ovar-
ian tumorigenesis (18), CSCs have been increasingly documented
in OC cell lines, and tumor tissues and serum of patients (712).
Using various markers characterizing ovarian CSCs such as CD44,
117, 133, 24, and aldehyde dehydrogenase-1 A1 (ALDH1), several
studies suggest that the central regulatory roles of miRs in OC
tumorigenesis are derived from their involvement in genetic alter-
ation of CSCs that affects the functionality of CSCs, as summarized
in Table 1.
Chen et al. compared the expression levels of miR-200c
between CD117+CD44+ovarian CSCs and CD117CD44
CSCs and found that miR-200c was present predominantly
in CD117CD44CSCs. Overexpression of miR-200c in
CD117+CD44+CSCs significantly down-regulated the expression
of ZEB-1 and vimentin, leading to an up-regulation of E-cadherin
www.frontiersin.org June 2014 | Volume 4 | Article 143 | 1
Wang et al. miRs in ovarian cancer metastasis
FIGURE 1 | microRNAs regulating tumor metastasis in ovarian cancer.
(A) Normal epithelium and potentially harbored stem cells. (B) miRs that are
implicated in modulation of cancer stem cell (CSC) transformation and
epithelial–mesenchymal transition (EMT) process. (C) miRs that are involved
in tumor angiogenesis. Tumor angiogenesis, which is required for further
tumor growth, is proven to be a handy process that facilitates tumor
metastasis. (D) miR-355 that increases cell–extracellular matrix (ECM)
interaction.
expression and dramatic reduction in the capacity of colony for-
mation, migration, and invasion in vitro. The miR-200c overex-
pression also decreased the magnitude of metastasis in xenograft
models of CD117+CD44+CSCs (7). Similarly, miR-200a was also
down-regulated in ovarian CD133+CSCs than in CD133CSCs,
and gain-of-function of miR-200a in CD133+cells inhibited the
migration and invasion of CD133+ovarian CSCs. Mechanisti-
cally, ZEB-2 was shown to be the target of miR-200a (8). Xu et al.
examined the role of miR-214 on formation of ovarian CSC phe-
notype, and found that enforced expression of miR-214 targeted
P53/Nanog axis and contributed to ovarian CSC confluence and
the ability of self-renewal (10).
microRNAs may also underlie the mechanism of CSC-induced
development of chemo-resistance. Park et al. showed overexpres-
sion of miR-23b, miR-27a, miR-27b, miR-346, miR-424, and miR-
503 in ALDH1+and chemoresistant OC cells, and among those,
miR-27 expression level was also correlated with metastasis mag-
nitude of OC (9). In human OC cells, miR-199a repressed CD44
expression and inhibited the proliferation, migration, and inva-
sion of CD44+CD117+ovarian CSCs. The inhibition of CD44 by
miR-199a reduced the expression of the multidrug resistance gene
ABCG2, and thereby increased the chemosensitivity of ovarian
CSCs (11). In addition, miR-199a was also implicated in cisplatin
resistance by which its inhibition increased mTOR expression and
decreased cisplatin-induced apoptosis in vitro (12).
microRNAs AND EPITHELIAL–MESENCHYMAL TRANSITION
Epithelial–mesenchymal transition in tumor is a process in which
tumor cells loosen various interactions including cell–cell and cell–
matrix, and are ready to detach, migrate, and metastasize through
blood or lymph vessels (13,21,23). EMT is thought to be a cru-
cial event that leads to tumor metastasis, possibly by increasing
stem-like properties in cancer cells and facilitating their seeding in
distance to form secondary tumors as illustrated in Figures 1A,B.
Accumulating data points to regulatory roles of miRs on EMT
phenotype, which is controlled by canonical pathways such as
WNT and transforming growth factor pathways in OC as shown
in Table 1.
microRNA-125a is widely reported as a tumor suppressor in
various malignancies including OC. Karen et al. showed that
miR-125a can inhibit EMT process in which overexpression of
miR-125a induced the reversal of EMT in highly invasive OC cells
(13). Using multiple biochemical approaches, epidermal growth
factor receptor (EGFR) was proposed as a signaling pathway to
suppress miR-125a expression via ETS family transcription fac-
tor, PEA3 in that study. Moreover, AT-rich interactive domain
3B (ARID3B) was shown to be the target of miR-125a to exert
the EMT repression function (13). Similarly, miR-138 was down-
regulated in highly invasive OC cell lines whereas in vivo overex-
pression of miR-138 inhibited OC metastasis (14). miR-138 has
been shown to directly repress the expression of genes associated
Frontiers in Oncology | Molecular and Cellular Oncology June 2014 | Volume 4 | Article 143 | 2
Wang et al. miRs in ovarian cancer metastasis
Table 1 | microRNAs aberrantly expressed in ovarian cancer and their target genes.
miRNA Targets Effects on tumor metastasis Reference
CSC-RELATED miRs
miR-23b N/I Enhances CSC transformation (9)
miR-27a, b VEGF, Cox2, and Sp1 Enhance CSC transformation and angiogenesis (9,19)
miR-214 P53/Nanog Enhances CSC transformation (10)
miR-346 N/I Enhances CSC transformation (9)
miR-424 N/I Enhances CSC transformation (9)
miR-503 N/I Enhances CSC transformation (9)
miR-199a CD44, mTOR, HIF-1α, VEGF, HER2, and HER3 Inhibits CSC transformation and angiogenesis (11,12,20)
miR-200a ZEB-2 Inhibits CSC transformation and EMT process (8,15)
miR-200c ZEB-1 and vimentin Inhibits CSC transformation and EMT process (7,15)
EMT-RELATED miRs
miR-20a PTEN Enhances EMT process (21)
miR-34 N/I Inhibits EMT process (22,23)
miR-125a AT-rich interactive domain 3B (ARID3B) Inhibits EMT process (13)
miR-138 SRY-related high-mobility group box (SOX4) and HIF-1αInhibits EMT process (14)
miR-141 N/I Inhibits EMT process (15)
miR-200b N/I Inhibits EMT process (15)
miR-429 N/I Inhibits EMT process (15)
ECM-RELATED miR
miR-355 Tenascin C (TNC) Increases cell–ECM interaction (24)
ANGIOGENESIS-RELATED miRs
miR-22 N/I Increases angiogenesis (25)
miR-150, 146a N/I Increase angiogenesis (26)
miR-182 BRCA1 Increases angiogenesis (27)
miR-124 N/I Decreases angiogenesis (28)
miR-125b HIF-1α, VEGF, HER2, and HER3 Decreases angiogenesis (20)
miR-145 p70S6K1, HIF-1α, and VEGF Decreases angiogenesis (29)
miR-484, 642, 217 VEGFB and VEGFR2 N/I (30)
N/I, not identified.
with EMT phenotype such as SRY-related high-mobility group
box (SOX4) and hypoxia-inducible factor-1α(HIF-1α). Accord-
ing to this study, loss of HIF-αdecreases slug expression through
proteasome-mediated degradation, whereas EGFR is inhibited by
knockdown of SOX4 (14).
In addition, miR-200 family including miR-200a, b, c, miR-141,
and miR-429 are all dysregulated in OC cells undergoing EMT.
For instance, the expression levels of miR-141 and miR-429 were
significantly lower in mesenchymal-like and more metastatic HEY
cells compared with epithelial-like and less invasive OVCAR3 cells.
Forced overexpression of miR-429 and miR-141 in HEY cells was
able to repress the signature characteristics of mesenchymal-like
cells but activate those of epithelial-like cells (15). This finding
indicates that miR-200 family not only inhibits CSC phenotyp-
ical formation but also acts as a gatekeeper to prevent OC cells
from detaching and intravasation. Interestingly, cancer-associated
miRs are often found in downstream of tumor suppressors or
oncogenes. For example, miR-34, a tumor suppressor, is trans-
activated by P53 (22). The expression of miR-34 is lower in OC
compared with normal tissue and is further down-regulated with
disease progression. Reconstitution of miR-34 results in decreased
proliferation, progression/invasion, and reversal of EMT (23).
Although most of miRs uncovered in ovarian EMT confer repres-
sive effects on EMT (i.e., MET), miR-20a has been shown to down-
regulate the expression of PTEN, a tumor suppressor and conse-
quently render ovarian CSC properties and EMT phenotypical
transformation (21).
microRNAs AND EXTRACELLULAR MATRIX MODIFICATION
Extracellular matrix (ECM) is composed of interlocking fibrous
proteins and glycosaminoglycans (GAGs), which provide struc-
tural and biochemical support for cells surrounded as illustrated
in Figure 1D. Tumors achieve invasion and metastasis through
modulating the microenvironment, in particular the ECM that
surrounds them (3133). Aberrant expression of miRs could ren-
der tumor cells metastatic capacity through destruction of ECM as
shown in Table 1. For example, in glioma, miR-21 was suggested
to be an oncomiR because of its ability to repress the expression
of RECK and TIMP3 which are the inhibitors of matrix met-
alloproteinase (MMPs), thereby conferring the metastatic capa-
bilities (34). In human hepatocellular cancer (HCC), miR-21
also repressed PTEN expression and consequently increased focal
adhesion kinase phosphorylation and expression of matrix MMP-
2 and 9 (35). In addition to the regulation of canonical MMPs,
www.frontiersin.org June 2014 | Volume 4 | Article 143 | 3
Wang et al. miRs in ovarian cancer metastasis
some miRs are involved in cell–ECM interactions and expression
of ECM components. The glycoprotein tenascin C (TNC), a pro-
tein that inhibits the interaction between cell–ECM, was shown to
be a target of miR-355, and loss of miR-355 potentiatedtumor cells’
metastatic abilities (31). miR-29c directly repressed the expres-
sion of ECM-related genes such as laminin gamma 1 and multiple
collagens in nasopharyngeal carcinomas (NPCs) (32).
While the involvement of miRs in ECM formation and mainte-
nance is increasingly reported in breast cancer, glioma, and HCC,
the study in OC is still emerging. Interestingly, Sorrentino et al.
demonstrated that miR-355, which has been shown to inhibit
tumor metastasis by targeting TNC expression (31,33,36) was
down-regulated in three OC cell lines resistant to paclitaxel and
one cell line resistant to cisplatin, suggesting potential connection
between ECM alteration and EMT transformation that can lead
to chemo-resistance in OC (24).
microRNAs AND ANGIOGENESIS
Tumor angiogenesis is the vital process through which tumors get
further progressed and transformed from benign states to malig-
nant states. It is contentious whether angiogenesis is necessary to
tumor metastasis, but this handy process during tumor progres-
sion facilitates cells to reach and disseminate via blood circulation
(19,20,29) as illustrated in Figure 1C. In response to hypoxic con-
ditions, tumor cells during progression increase the expression of
miRs that sustain tumor growth and neo-angiogenesis, or decrease
the expression of miRs that repress neo-angiogenesis. For exam-
ple, the up-regulated miR-27a is able to inhibit ZBTB10 expression
and thereby indirectly regulates the expression levels of vascular
endothelial growth factor (VEGF) and VEGF receptor (VEGFR).
Conversely, miR-16, miR-15b, miR-20a, and miR-20b that repress
angiogenesis through modulation of VEGF and VEGFR levels are
consistently down-regulated in tumor cells. More recent studies
shed light on the roles of miRs in OC angiogenesis as summarized
in Table 1.
Xu et al. has shown that miR-145 was repressed in OC tis-
sues and cell lines due to its inhibitory effect on neo-angiogenesis
(29). A mechanistic study demonstrated that miR-145 was able to
repress p70S6K1 expression and thereby inhibited the expression
of both HIF-1αand VEGF. Similarly, miR-125b and miR-199a
were shown to be tumor-suppressive miRs by targeting HIF-1α
and VEGF and consequently reduced angiogenesis (20). In addi-
tion, HER2 and HER3 were suggested to be the direct targets of
miR-125b and miR-199a because overexpression of miR-125b and
miR-199a led to the failure of angiogenesis in xenograft mod-
els of OC, which is mediated by these two genes. Some miRs
such as miR-217, miR-484, and miR-642 have been shown to
directly modulate VEGFB and VEGFR2 pathways and predict
tumor chemo-resistance (30). Ablation of miR-27a also repressed
the expression of VEGF as well as Cox2 and Sp1. In particular,
miR-27a appears to play a central role in follicle-stimulating
hormone (FSH)-mediated angiogenesis in OC (19).
CIRCULATING microRNAs IN OVARIAN CANCER
Ovarian cancer is the most fatal gynecologic malignancy among
women in the Unites States. Current treatment is based on surgery
in combination with platinum-based chemotherapy. However,
once patients with OC are diagnosed at advanced stages, there
is limited availability of effective treatments. Therefore, the dis-
covery and adoption of early detection biomarker is critical for
the improved outcome. Cancer antigen 125 (CA-125) is the most
commonly used marker to detect OC, monitor the response of
OC to treatment, and predict patients’ prognosis after treatment.
However, due to its lack of sensitivity, CA-125 is only detectable
when patients are advanced to late stages. Furthermore, serum
CA-125 levels are non-specifically elevated in patients with other
cancers including cancers of lung, breast, and gastrointestinal
tract (26).
Lawrie et al. first described that the serum levels of miR-21
were correlated with relapse-free survival in patients with diffuse
large B-cell lymphoma (37). Subsequently, serum miR-141 levels
have been used to monitor patients with prostate cancer. Using
mouse prostate cancer xenograft models, the presence of tumor-
derived miR-141 in mouse serum was confirmed (38). Chen et al.
compared miR expression signature in patients with lung and col-
orectal cancer with healthy subjects and found that miR-25 and
miR-223 were overexpressed in cancer patients (39,40). Circulat-
ing miRs were also detected in patients with HCC, non-small-cell
lung cancer (NSCLC), prostate cancer, breast cancer, and gas-
tric cancer (41). Among patients with OC, miR-21, miR-29a,
miR-92, miR-93, miR-99b, miR-126, miR-127, and miR-155 were
aberrantly expressed in blood compared to healthy controls (40).
Because miRs were detected in patients’ serum even at early
stage of various cancers, miR signatures of tumor-derived exo-
somes or other microvesicles have been proposed as diagnostic
biomarkers of many cancers (4244). Unlike those synthetic RNAs
that are degraded by RNases in blood, miRs are incorporated
into a membrane-enclosed complex and resistant to RNases in
plasma. In addition, the levels of miRs in serum have been shown
to be particularly stable against temperature and pH changes,
rendering miRs as reliable diagnostic, prognostic, and predictive
biomarkers (39,45).
CONCLUSION AND PERSPECTIVE
Metastasis is a multifactorial complex process in which tumor
cells gain CSC-like properties and undergo EMT phenotypical
changes through degrading ECM to disseminate into non-adjacent
tissues with bloodstream. microRNAs play central roles in initia-
tion, progression, invasion, and metastasis of cancer. While most
miR signatures in cancers are involved in both tumor growth and
metastasis, there are a few groups of them that were important for
tumor metastasis, while exerting no effects on tumor proliferation
(46). For example, miR-22 has just been linked to progression of
OC to late stages, but no effects on cell viability and apoptosis (25).
miR-150 and miR-146a are highly expressed in omental lesion
compared to primary tumor sites and enhance spheroid forma-
tion to promote OC metastasis and chemoresistance to cisplatin
(26). In addition, miR-124 is down-regulated in clinical OC speci-
mens compared to adjacent normal tissues and correlates with the
severity of metastasis both in vivo and in vitro (28). miR-182 is
shown to be oncogenic, particularly in high-grade serous ovarian
carcinoma (HG-SOC). miR-182 targets breast cancer 1 (BRCA1)
and metastasis suppressor 1 (MTSS1) to impair the repair of DNA
double-strand breaks, but simultaneously enhances the expression
Frontiers in Oncology | Molecular and Cellular Oncology June 2014 | Volume 4 | Article 143 | 4
Wang et al. miRs in ovarian cancer metastasis
of high-mobility group AT-hook2 (HMGA2) (27). Taken together,
it is well-appreciated that miRs are able to alter metastasis status
by modulating the expression of genes involved in CSC transfor-
mation, EMT, angiogenesis, and ECM as shown in Table 1 and
Figure 1. As we also reviewed the research on circulating miRs,
miRs can be promising biomarkers for OC. Further studies are
warranted to investigate their potential diagnostic and therapeutic
utilities for patients with OC.
ACKNOWLEDGMENTS
We thank editors for inviting us to write this mini-review. Due to
space restrictions, the authors cannot cite many important liter-
atures in this field. The authors apologize to all colleagues whose
work contributed significantly. This work was supported by Geor-
gia Regents University Departmental Start-up Fund andAmerican
Heart Association Grant-in-Aid 12GRNT12100048 and Scientist
Development Grant 14SDG18970040 to Il-man Kim.
REFERENCES
1. Siegel R, DeSantis C, Virgo K, Stein K, Mariotto A, Smith T, et al. Cancer treat-
ment and survivorship statistics, 2012. CA Cancer J Clin (2012) 62(4):220–41.
doi:10.3322/caac.21149
2. Nossov V, Amneus M, Su F, Lang J, Janco JM, Reddy ST, et al. The early detec-
tion of ovarian cancer: from traditional methods to proteomics. Can we really
do better than serum CA-125? Am J Obstet Gynecol (2008) 199(3):215–23.
doi:10.1016/j.ajog.2008.04.009
3. Bast RC Jr, Xu FJ, Yu YH, Barnhill S, Zhang Z, Mills GB. CA 125: the past and
the future. Int J Biol Markers (1998) 13(4):179–87.
4. Bagan P,Berna P,Assouad J, HupertanV, Le Pimpec Barthes F, Riquet M. Value of
cancer antigen 125 for diagnosis of pleural endometriosis in females with recur-
rent pneumothorax. Eur Respir J (2008) 31(1):140–2. doi:10.1183/09031936.
00094206
5. Sarandakou A, Protonotariou E, Rizos D. Tumor markers in biological flu-
ids associated with pregnancy. Crit Rev Clin Lab Sci (2007) 44(2):151–78.
doi:10.1080/10408360601003143
6. Asher V, Hammond R, Duncan TJ. Pelvic mass associated with raised CA
125 for benign condition: a case report. World J Surg Oncol (2010) 8:28.
doi:10.1186/1477-7819- 8-28
7. Chen D, Zhang Y, Wang J, Chen J, Yang C, Cai K, et al. microRNA-200c over-
expression inhibits tumorigenicity and metastasis of CD117+CD44+ ovarian
cancer stem cells by regulating epithelial-mesenchymal transition. J Ovarian Res
(2013) 6(1):50. doi:10.1186/1757-2215-6-50
8. Wu Q,Guo R, Lin M, Zhou B, Wang Y. microRNA-200a inhibits CD133/1+ ovar-
ian cancer stem cells migration and invasion by targeting E-cadherin repressor
ZEB2. Gynecol Oncol (2011) 122(1):149–54. doi:10.1016/j.ygyno.2011.03.026
9. Park YT, Jeong JY, Lee MJ, Kim KI, Kim TH, Kwon YD, et al. microRNAs over-
expressed in ovarian ALDH1-positive cells are associated with chemoresistance.
J Ovarian Res (2013) 6(1):18. doi:10.1186/1757-2215-6-18
10. Xu CX, Xu M, Tan L, Yang H, Permuth-Wey J, Kruk PA, et al. microRNA miR-
214 regulates ovarian cancer cell stemness by targeting p53/Nanog. J Biol Chem
(2012) 287(42):34970–8. doi:10.1074/jbc.M112.374611
11. Cheng W, Liu T, Wan X, Gao Y, Wang H. microRNA-199a targets CD44 to sup-
press the tumorigenicity and multidrug resistance of ovarian cancer-initiating
cells. FEBS J (2012) 279(11):2047–59. doi:10.1111/j.1742-4658.2012.08589.x
12. Wang Z, Ting Z, Li Y, Chen G, Lu Y, Hao X. microRNA-199a is able to
reverse cisplatin resistance in human ovarian cancer cells through the inhi-
bition of mammalian target of rapamycin. Oncol Lett (2013) 6(3):789–94.
doi:10.3892/ol.2013.1448
13. Cowden Dahl KD, Dahl R,Kruichak JN, Hudson LG. The epidermal growth fac-
tor receptor responsive miR-125a represses mesenchymal morphology in ovar-
ian cancer cells. Neoplasia (2009) 11(11):1208–15. doi:10.1593/neo.09942
14. Yeh YM, Chuang CM, Chao KC, Wang LH. microRNA-138 suppresses ovar-
ian cancer cell invasion and metastasis by targeting SOX4 and HIF-1alpha. Int
J Cancer (2013) 133(4):867–78. doi:10.1002/ijc.28086
15. Jabbari N, Reavis AN, McDonald JF. Sequence variation among members of the
miR-200 microRNA family is correlated with variation in the ability to induce
hallmarks of mesenchymal-epithelial transition in ovarian cancer cells. J Ovar-
ian Res (2014) 7(1):12. doi:10.1186/1757-2215- 7-12
16. Clevers H. The cancer stem cell: premises, promises and challenges. Nat Med
(2011) 17(3):313–9. doi:10.1038/nm.2304
17. Baccelli I, Trumpp A. The evolving concept of cancer and metastasis stem cells.
J Cell Biol (2012) 198(3):281–93. doi:10.1083/jcb.201202014
18. Bapat SA, Mali AM, Koppikar CB, Kurrey NK. Stem and progenitor-like cells
contribute to the aggressive behavior of human epithelial ovarian cancer.Cancer
Res (2005) 65(8):3025–9. doi:10.1158/0008-5472.CAN-04- 3931
19. Lai Y,Zhang X, Zhang Z, ShuY,Luo X, Yang Y, et al. The microRNA-27a: ZBTB10-
specificity protein pathway is involved in follicle stimulating hormone-induced
VEGF,Cox2 and survivin expression in ovarian epithelial cancer cells. Int J Oncol
(2013) 42(2):776–84. doi:10.3892/ijo.2012.1743
20. He J, Jing Y, Li W, Qian X, Xu Q, Li FS, et al. Roles and mechanism of miR-
199a and miR-125b in tumor angiogenesis. PLoS One (2013) 8(2):e56647.
doi:10.1371/journal.pone.0056647
21. Luo X, Dong Z, Chen Y, Yang L, Lai D. Enrichment of ovarian cancer stem-like
cells is associated with epithelial to mesenchymal transition through an miRNA-
activated AKT pathway. Cell Prolif (2013) 46(4):436–46. doi:10.1111/cpr.12038
22. Chang TC, Wentzel EA, Kent OA, Ramachandran K, Mullendore M, Lee KH,
et al. Transactivation of miR-34a by p53 broadly influences gene expression and
promotes apoptosis. Mol Cell (2007) 26(5):745–52. doi:10.1016/j.molcel.2007.
05.010
23. Corney DC, Hwang CI, Matoso A, Vogt M, Flesken-Nikitin A, Godwin AK, et al.
Frequent downregulation of miR-34 family in human ovarian cancers. Clin
Cancer Res (2010) 16(4):1119–28. doi:10.1158/1078-0432.CCR-09-2642
24. Sorrentino A, Liu CG, Addario A, Peschle C, Scambia G, Ferlini C. Role
of microRNAs in drug-resistant ovarian cancer cells. Gynecol Oncol (2008)
111(3):478–86. doi:10.1016/j.ygyno.2008.08.017
25. Li J, Liang S, Yu H, Zhang J, Ma D, Lu X. An inhibitory effect of miR-22 on cell
migration and invasion in ovarian cancer. Gynecol Oncol (2010) 119(3):543–8.
doi:10.1016/j.ygyno.2010.08.034
26. Vang S, Wu HT, Fischer A, Miller DH, MacLaughlan S, Douglass E, et al. Iden-
tification of ovarian cancer metastatic miRNAs. PLoS One (2013) 8(3):e58226.
doi:10.1371/journal.pone.0058226
27. Liu Z, Liu J, Segura MF, Shao C, Lee P, Gong Y, et al. miR-182 overexpres-
sion in tumourigenesis of high-grade serous ovarian carcinoma. J Pathol (2012)
228(2):204–15. doi:10.1002/path.4000
28. Zhang H, Wang Q, Zhao Q, Di W. miR-124 inhibits the migration and inva-
sion of ovarian cancer cells by targeting SphK1. J Ovarian Res (2013) 6(1):84.
doi:10.1186/1757-2215- 6-84
29. Xu Q, Liu LZ, Qian X, Chen Q, Jiang Y, Li D, et al. miR-145 directly targets
p70S6K1 in cancer cells to inhibit tumor growth and angiogenesis. Nucleic Acids
Res (2012) 40(2):761–74. doi:10.1093/nar/gkr730
30. Vecchione A, Belletti B, Lovat F, Volinia S, Chiappetta G, Giglio S, et al. A
microRNA signature defines chemoresistance in ovarian cancer through mod-
ulation of angiogenesis. Proc Natl Acad Sci U S A (2013) 110(24):9845–50.
doi:10.1073/pnas.1305472110
31. Orend G, Chiquet-Ehrismann R. Tenascin-C induced signaling in cancer. Cancer
Lett (2006) 244(2):143–63. doi:10.1016/j.canlet.2006.02.017
32. Sengupta S, den Boon JA, Chen IH, Newton MA, Stanhope SA, Cheng YJ, et al.
microRNA 29c is down-regulated in nasopharyngeal carcinomas, up-regulating
mRNAs encoding extracellular matrix proteins. Proc Natl Acad Sci U S A (2008)
105(15):5874–8. doi:10.1073/pnas.0801130105
33. Negrini M, Calin GA. Breast cancer metastasis: a microRNA story. Breast Cancer
Res (2008) 10(2):203. doi:10.1186/bcr1867
34. Gabriely G, Wurdinger T, Kesari S, Esau CC, Burchard J, Linsley PS, et al.
microRNA 21 promotes glioma invasion by targeting matrix metalloproteinase
regulators. Mol Cell Biol (2008) 28(17):5369–80. doi:10.1128/MCB.00479-08
35. Meng F,Henson R, Wehbe-Janek H,Ghoshal K, Jacob ST,Patel T. microRNA-21
regulates expression of the PTEN tumor suppressor gene in human hepatocel-
lular cancer. Gastroenterology (2007) 133(2):647–58. doi:10.1053/j.gastro.2007.
05.022
36. Tavazoie SF, Alarcon C, Oskarsson T, Padua D, Wang Q, Bos PD, et al. Endoge-
nous human microRNAs that suppress breast cancer metastasis. Nature (2008)
451(7175):147–52. doi:10.1038/nature06487
www.frontiersin.org June 2014 | Volume 4 | Article 143 | 5
Wang et al. miRs in ovarian cancer metastasis
37. Lawrie CH, Gal S, Dunlop HM, Pushkaran B, Liggins AP, Pulford K, et al.
Detection of elevated levels of tumour-associated microRNAs in serum of
patients with diffuse large B-cell lymphoma. Br J Haematol (2008) 141(5):672–5.
doi:10.1111/j.1365-2141.2008.07077.x
38. Mitchell PS, Parkin RK, Kroh EM, Fritz BR,Wyman SK, Pogosova-Agadjanyan
EL, et al. Circulating microRNAs as stable blood-based markers for cancer
detection. Proc Natl Acad Sci U S A (2008) 105(30):10513–8. doi:10.1073/pnas.
0804549105
39. Chen X, Ba Y, Ma L, Cai X, Yin Y, Wang K, et al. Characterization of microRNAs
in serum: a novel class of biomarkers for diagnosis of cancer and other diseases.
Cell Res (2008) 18(10):997–1006. doi:10.1038/cr.2008.282
40. Resnick KE, Alder H, Hagan JP, Richardson DL, Croce CM, Cohn DE. The
detection of differentially expressed microRNAs from the serum of ovarian
cancer patients using a novel real-time PCR platform. Gynecol Oncol (2009)
112(1):55–9. doi:10.1016/j.ygyno.2008.08.036
41. Hu Z, Chen X, Zhao Y, Tian T, Jin G, Shu Y, et al. Serum microRNA signa-
tures identified in a genome-wide serum microRNA expression profiling pre-
dict survival of non-small-cell lung cancer. J Clin Oncol (2010) 28(10):1721–6.
doi:10.1200/JCO.2009.24.9342
42. Valadi H, Ekstrom K, Bossios A, Sjostrand M, Lee JJ, Lotvall JO. Exosome-
mediated transfer of mRNAs and microRNAs is a novel mechanism of
genetic exchange between cells. Nat Cell Biol (2007) 9(6):654–9. doi:10.1038/
ncb1596
43. Rabinowits G, Gercel-Taylor C, Day JM, Taylor DD, Kloecker GH. Exosomal
microRNA: a diagnostic marker for lung cancer. Clin Lung Cancer (2009)
10(1):42–6. doi:10.3816/CLC.2009.n.006
44. Taylor DD, Gercel-Taylor C. microRNA signatures of tumor-derived exosomes
as diagnostic biomarkers of ovarian cancer. Gynecol Oncol (2008) 110(1):13–21.
doi:10.1016/j.ygyno.2008.04.033
45. Kroh EM, Parkin RK, Mitchell PS,Tewari M. Analysis of circulating microRNA
biomarkers in plasma and serum using quantitative reverse transcription-PCR
(qRT-PCR). Methods (2010) 50(4):298–301. doi:10.1016/j.ymeth.2010.01.032
46. Ma L, Teruya-Feldstein J, Weinberg RA. Tumour invasion and metastasis ini-
tiated by microRNA-10b in breast cancer. Nature (2007) 449(7163):682–8.
doi:10.1038/nature06174
Conflict of Interest Statement: The authors declare that the researchwas conducted
in the absence of any commercial or financial relationships that could be construed
as a potential conflict of interest.
Received: 21 April 2014; paper pending published: 19 May 2014; accepted: 27 May
2014; published online: 10 June 2014.
Citation: Wang Y, Kim S and Kim I (2014) Regulation of metastasis by microRNAs in
ovarian cancer. Front. Oncol. 4:143. doi: 10.3389/fonc.2014.00143
This article was submitted to Molecular and Cellular Oncology, a section of the journal
Frontiers in Oncology.
Copyright © 2014 Wang, Kim and Kim. This is an open-access article distributed under
the terms of the Creative Commons Attribution License (CC BY). The use, distribution
or reproduction in other forums is permitted, provided the original author(s) or licensor
are credited and that the original publication in this journal is cited, in accordance with
accepted academic practice. No use, distribution or reproduction is permitted which
does not comply with these terms.
Frontiers in Oncology | Molecular and Cellular Oncology June 2014 | Volume 4 | Article 143 | 6
... MicroRNAs (miRNAs/miRs), ~22 nucleotides, exert crucial roles via inhibiting translation or promoting degradation (11,12). Multiple studies demonstrated that miRNAs participated in the progression of various cancer types, including lung cancer (13), osteosarcoma (14), thyroid cancer (15) and OC (16). ...
Article
Full-text available
Long non-coding RNAs (lncRNAs) and microRNAs (miRNAs/miRs) were reported to be associated with the development of ovarian cancer (OC). Increasing evidence demonstrated that lncRNA SNHG20 and miR-338-3p were involved in OC. However, the functional mechanism of lncRNA SNHG20 and miR-338-3p in OC development remains unknown. The expression of SNHG20, miR-338-3p and myeloid cell leukemia 1 (MCL1) was detected by reverse transcription-quantitative PCR. MTT assay, flow cytometry and transwell migration and invasion assays were used to assess cell proliferation, apoptosis, migration and invasion, respectively. The relative protein expression was detected by western blot analysis. The interaction between miR-338-3p and SNHG20 or MCL1 was predicted by starBase v3.0, and subsequently confirmed by dual-luciferase reporter assay. Besides, mouse xenograft assay was carried out to explore the effect of SNHG20 on tumor growth in vivo. The levels of SNHG20 and MCL1 were upregulated, while miR-338-3p level was downregulated in OC tissues and cells. SNHG20 knockdown repressed OC cell proliferation, migration, invasion and epithelial-mesenchymal transition, and induced apoptosis. Interestingly, SNHG20 targeted miR-338-3p to regulate MCL1 expression. miR-338-3p depletion or MCL1 overexpression could reverse the effects of SNHG20 knockdown on OC cells. Besides, SNHG20 knockdown impeded tumor growth in vivo. In conclusion, the present study demonstrated that SNHG20 regulates OC development via modulation of the miR-338-3p/MCL1 axis, providing the theoretical basis for the treatment of OC.
... There are also miRNAs with opposing roles in EMT and chemoresistance wherein they promote such mechanisms e.g. miR-20a which induces cisplatin resistance and EMT in OVCAR3 138,139 . Overexpression of miR-181a induces EMT and resistance to paclitaxel in SKOV3 cells through overexpression of P-glycoprotein 140 . ...
Article
Full-text available
Cellular plasticity, by large, is the ability through which cells morph into new phenotypic identity by trading-off with the previous one. This phenomenon has been observed in the normal and tumor cells in a very similar fashion. Occurrence of cellular plasticity, in malignancies like epithelial ovarian carcinoma (EOC) are well known but highly debated in terms of origin of the tumor for the inherent heterogeneity across subtypes. EOC has been termed as a clinically challenging malady for its subversive nature against chemotherapy. The management of ovarian cancer is mostly hindered by relapse with recalcitrance towards primary chemotherapy regimen e.g. platinum–taxol combination. Also, late detection preceded by peritoneal metastasis poses another challenge to the treatment. Underlying both these aspects of the disease, tumor heterogeneity turns out as the most critical factor. In the light of heterogeneity that can be across patients (inter-tumoral) and/or within a tumor (intra-tumoral), ovarian carcinoma is a multifactorial ailment. The governing factors behind this heterogeneity are—diverse genetic landscape among subtypes of EOC; the presence of cancer stem cell (CSC) niche and inherent plasticity of the cancer cells themselves. Apart from the well-studied mechanisms of plasticity, there are several emerging molecular players like lncRNAs, Hippo pathway and also phenomena like dedifferentiation of non-CSC neoplastic cells ,and transdifferentiation of CSCs. Here, we present an overview of the current knowledge on the evolution of EOC through cellular plasticity with emphasis on the three major aspects namely, subtype-specific genetic diversity; ovarian CSC and cancer cell duality between epithelial and mesenchymal lineages.
... miRNAs directly bind to the 3'untranslated region (3'UTR) of mRNAs, resulting in translational repression or mRNA degradation (10). Mounting evidence has revealed that miRNAs are involved in various biological processes (11)(12)(13), including cell growth, cell apoptosis and cell differentiation (11,(14)(15)(16)(17)(18)(19). Altered expression of various miRNAs following traumatic SCI has been observed in adult rats (20). ...
Article
Full-text available
MicroRNAs (miRs) play an important role in the development and progression of spinal cord injury (SCI). The role of miR‑138‑5p in SCI was investigated in the present study. The anti‑inflammatory effects of miR‑138‑5p and underlying mechanisms were investigated in an SCI rat model and in vitro model. Reverse transcription‑quantitative PCR (RT‑qPCR) was used to examine the expression of miR‑138‑5p in the SCI in vivo and in vitro models, as well as patients with SCI; it was found that miR‑138‑5p was significantly upregulated in SCI. Bioinformatics and dual‑luciferase reporter assays were performed to predict and confirm the binding sites between miR‑138‑5p and the 3'untranslated region of sirtuin 1 (SIRT1). Then, the expression of SIRT1 was detected via RT‑qPCR and western blotting, indicating downregulation of SIRT1 in SCI. PC12 cells were transfected with miR‑138‑5p inhibitor, inhibitor control or miR‑138‑5p inhibitor + SIRT1 small interfering RNA for 48 h, and then subjected to lipopolysaccharide (100 ng/ml) treatment for 4 h. Then, MTT assay, flow cytometry and ELISA experiments were performed to analyze cell viability, apoptosis, and the levels of tumor necrosis factor‑α, interleukin (IL)‑1β and IL‑6. Findings suggested that downregulation of miR‑138‑5p increased PC12 cell viability, inhibited cell apoptosis and attenuated proinflammatory responses, which may result in amelioration of SCI. However, all these effects were reversed by SIRT1 knockdown. Finally, it was observed that miR‑138‑5p altered the related protein expression of the PTEN/AKT pathway. These results indicated that miR‑138‑5p could regulate inflammatory responses and cell apoptosis in SCI models by modulating the PTEN/AKT signaling pathway via SIRT1, thus playing an important role in the development of SCI. Collectively, the present study demonstrated that miR‑138‑5p may be a novel therapeutic target for the treatment of SCI.
... The role of miRNAs in the initiation, growth and progress has already been identified. 34,35 miR-363, a cancer-related miRNA, plays an oncogenic as well as tumor suppressor role in many cancers. miR-363 has been found to play a tumor-suppressor role in OC by inhibiting NIN1/ RPN12 binding protein 1 homolog (NOB1). ...
... The role of miRNAs in the initiation, growth and progress has already been identified. 34,35 miR-363, a cancer-related miRNA, plays an oncogenic as well as tumor suppressor role in many cancers. miR-363 has been found to play a tumor-suppressor role in OC by inhibiting NIN1/ RPN12 binding protein 1 homolog (NOB1). ...
Article
Full-text available
MicroRNAs, are regulatory small non-coding RNAs, which have recently been identified for the post-transcriptional regulation of gene expression in a variety of cellular processes especially important roles in disease and tissue remodeling. Apart from involvement in a variety of biological processes, microRNAs were early recognized for their potential use as biomarkers in disease diagnostics and therapeutics. Currently, there are number of microRNAs helping clinicians to determine the origins of cancer and other diseases. Though more than a decade of research on miRNA, yet the potency of their use is still in progress. The development of microRNA therapeutics has proved more challenging mainly due to delivery issues. This review article mainly focuses on the emerging insights involving miRNA biogenesis and their role as clinical diagnostic and therapeutic non-invasive biomarkers in various pathological conditions such as cancer, neurodegenerative disorders, polycystic ovarian syndrome and diabetes mellitus.
Article
Full-text available
Ovarian cancer is one of the most aggressive and significant malignant tumor forms in the female reproductive system. It is the leading cause of death among gynecological cancers owing to its metastasis. Since its preliminary disease symptoms are lacking, it is imperative to develop early diagnostic biomarkers to aid in treatment optimization and personalization. In this vein, microRNAs, which are short sequence non-coding molecules, displayed great potential as highly specific and sensitive biomarker. miRNAs have been extensively advocated and proven to serve an instrumental part in the clinical management of cancer, especially ovarian cancer, by promoting the cancer cell progression, invasion, delayed apoptosis, epithelial-mesenchymal transition, metastasis of cancer cells, chemosensitivity and resistance and disease therapy. Here, we cover our present comprehension of the most up-to-date microRNA-based approaches to detect ovarian cancer, as well as current diagnostic and treatment strategies, the role of microRNAs as oncogenes or tumor suppressor genes, and their significance in ovarian cancer progression, prognosis, and therapy
Article
HYOU1 encodes a protein from the endoplasmic reticulum chaperone proteins, expressed to protect cellular mechanisms from stress such as hypoxia, insufficient energy and excessive or insufficient substances, and to restore cell homeostasis. In this study, we report a novel pathogenic variant in HYOU1. The proband, the second patient with pathogenic variant in HYOU1, was a female born to consanguineous parents. A novel homozygous pathogenic variant in HYOU1 (NM_001130991.3: c.1456C>T; p.Arg486Cys) was identified, causing anemia, thrombocytopenia and severe panleukopenia and immunodeficiency in the second month of age, leading to consistent high-grade fever, regression of brain functions and recurrent infections; ultimately resulting in the patient expiring at three and half months of age. Both parents are heterozygous for this variant and have no issues related to this study.
Chapter
Cancer is one of the leading causes of death worldwide. Recent report from the World Health Organization suggested that, globally, one in six deaths is owing to cancer. In 2018, it was accountable for nearly 9.6 million deaths, and it is expected to be 14.6 million by the year 2035. The worldwide burden of cancer increase is due to aging and growth of population. In addition, cancer-associated lifestyle choices like smoking, sedentary habits and westernized diets increases the risk. Metastasis is complex and multistep process that results in the spread of cancerous cells from the primary site of the tumor to the surrounding tissues and to distant organs. Metastatic cancer is the primary cause of cancer morbidity and mortality. Several studies suggest that tumor has heterogeneous cell population and have numerically less cancer stem cell (CSC) population with self-renewal characteristics. CSCs are shown to drive tumor initiation, progression, metastasis, recurrence, and resistance. In addition, acquisition of epithelial-mesenchymal transition, expression of aberrant RNA-binding proteins, dysregulated microRNA expression, and increase in intercellular transfer of molecules via exosome cargo have been correlated with tumor progression, invasion, metastasis, poor survival, and an increased risk of cancer recurrence. Given the tumor initiating capacity, resistance, migratory potential and invasiveness, CSCs are the seeds of metastasis. This review article attempts to provide the details of the critical importance of CSCs on metastatic process and to offer a basis for the investigation of novel targets to curtail this deadly disease.
Article
During malignant transformation and cancer progression, tumor cells face both intrinsic and extrinsic stress, endoplasmic reticulum (ER) stress in particular. To survive and proliferate, tumor cells use multiple stress response pathways to mitigate ER stress, promoting disease aggression and treatment resistance. Among the stress response pathways is ER-associated degradation (ERAD), which consists of multiple components and steps working together to ensure protein quality and quantity. In addition to its established role in stress responses and tumor cell survival, ERAD has recently been shown to regulate tumor immunity. Here we summarize current knowledge on how ERAD promotes protein degradation, regulates immune cell development and function, participates in antigen presentation, exerts paradoxical roles on tumorigenesis and immunity, and thus impacts current cancer therapy. Collectively, ERAD is a critical protein homeostasis pathway intertwined with cancer development and tumor immunity. Of particular importance is the need to further unveil ERAD’s enigmatic roles in tumor immunity to develop effective targeted and combination therapy for successful treatment of cancer.
Article
Full-text available
Exosomes are vesicles of endocytic origin released by many cells. These vesicles can mediate communication between cells, facilitating processes such as antigen presentation. Here, we show that exosomes from a mouse and a human mast cell line (MC/9 and HMC-1, respectively), as well as primary bone marrow-derived mouse mast cells, contain RNA. Microarray assessments revealed the presence of mRNA from approximately 1300 genes, many of which are not present in the cytoplasm of the donor cell. In vitro translation proved that the exosome mRNAs were functional. Quality control RNA analysis of total RNA derived from exosomes also revealed presence of small RNAs, including microRNAs. The RNA from mast cell exosomes is transferable to other mouse and human mast cells. After transfer of mouse exosomal RNA to human mast cells, new mouse proteins were found in the recipient cells, indicating that transferred exosomal mRNA can be translated after entering another cell. In summary, we show that exosomes contain both mRNA and microRNA, which can be delivered to another cell, and can be functional in this new location. We propose that this RNA is called " exosomal shuttle RNA " (esRNA). Exosomes are small (50–90 nm) membrane vesicles of endocytic origin that are released into the extracellular environment on fusion of multivesicular bodies (MVB) with the plasma membrane 1. Many cells have the capacity to release exosomes, including reticulo-cytes 2 , dendritic cells 3 , B cells 4 , T cells 5 , mast cells 6 , epithelial cells 7 and tumour cells 8. The functions of exosomes are not completely understood, although it has been shown that exosomes can participate in the signalling events contributing to antigen presentation to T cells 4 and the development of tolerance 9. Several mechanisms have been hypothesized describing the interactions of exosomes and recipient cells. Exosomes can bind to cells through recep-tor–ligand interactions, similar to cell–cell communication mediating , for example, antigen presentation 4. Alternatively, exosomes could putatively attach or fuse with the target-cell membrane, delivering exosomal surface proteins and perhaps cytoplasm to the recipient cell 10,11. Finally, exosomes may also be internalized by the recipient cells by mechanisms such as endocytosis 12. Exosomes were isolated from a mast-cell line (MC/9), primary bone marrow-derived mast cells (BMMC) and a human mast-cell line (HMC-1) through a series of microfiltration and ultracentrifugation steps modified from what has been previously described 4. To confirm that the structures studied indeed are exosomes, they were examined by electron microscopy (Fig. 1a), flow cytometric analysis (FACS; Fig. 1b), and proteomic analysis (see Supplementary Information, Table S1). The electron micrographs of the exosomes revealed rounded structures with a size of approximately 50–80 nm, similar to previously described exo-somes 4,13–15. The identity of the studied vesicles was further confirmed as exosomes by FACS analysis (Fig. 1b), which show the presence of the surface protein CD63 — a commonly used marker of exosomes. Finally, extensive protein analysis of the MC/9 exosomes was performed on multiple samples using LC-MS/MS technology. A total of 271 proteins were identified (see Supplementary Information, Table S1) from three preparations of the isolated vesicles, of which 47 proteins were present in all three samples. More importantly, a large number of the proteins found in the preparations were the same as proteins previously identified in exosomes produced by other cells (that is, exosomes derived from intestinal epithelial cells, urine, dendritic cells, microglia, melanoma, T-cells and B-cells). In particular, 60% of the 47 proteins found in all samples of mast-cell exosomes have been previously found in other types of exosomes. Moreover, 39% of the 271 total proteins found in the analysed exosome samples have also been previously found in other types of exosomes. Thus, the electron microscopy, the FACS, and the detailed protein analyses each provided significant evidence in favour of the identification of the isolated vesicles as exosomes. The presence of nucleic acids was examined in exosomes derived from MC/9, BMMC and HMC-1 cells to define a potential mechanism by which exosomes may mediate cell–cell communication. These assessments showed that isolated exosomes contain no DNA (see Supplementary Information, Fig. S1). However, substantial amounts of RNA were detected by agarose gel electrophoresis, spectrophotometry
Article
Full-text available
Epithelial-Mesenchymal Transition (EMT) is a transient and reversible (Mesenchymal-Epithelial Transition or MET) process by which epithelial cells acquire mesenchymal cell characteristics including reduced intercellular adhesion and increased cell motility. While EMT/MET has long been recognized as an essential component of early embryonic development, there is a growing body of evidence indicating that EMT/MET is also a key component of ovarian cancer (OC) metastasis. Recent findings have implicated members of the miR-200 family of microRNAs (miRNAs) in this process. Individual members of the miR-200 family of miRNAs were transiently over expressed in metastatic (mesenchymal-like) OC cell lines. Changes in morphology, molecular profiles and drug sensitivity were monitored relative to cells transfected with a negative control. Morphological hallmarks of MET were detected in cells transfected with all miR-200 family members. Gene expression profiling demonstrated up regulation of epithelial biomarkers and down regulation of mesenchymal biomarkers in transfected cells although significant variation in molecular response and drug sensitivity was associated with different members of the miR-200 family. Our results indicate that although ectopic overexpression of all members of the miR-200 family in mesenchymal-like OC cells results in morphological changes characteristic of MET, the underlying molecular changes and induced drug sensitivities are highly variable and correlated with sequence variation within the seed and non-seed regions of individual family members.
Article
Full-text available
Epithelial ovarian cancer (EOC) is still a major gynecologic problem with poor 5 year survival rate due to distance metastases, despite routine surgery and chemotherapy. The precise underlying molecular mechanisms that trigger EOC migration and invasion are unclear. Recent studies suggest that the expression of microRNAs is widely dysregulated in ovarian cancer; and that they have evolved into tumorigenic processes, including cell proliferation, apoptosis and motility. The expression of miR-124 was assessed in clinical ovarian cancer specimens and cell lines using miRNA qRTPCR. The function of miR-124 on cell migration and invasion was confirmed in vitro through wound healing assay and transwell assay. Luciferase reporter assay was used to confirm target associations. We showed that miR-124 is down-regulated in ovarian cancer specimens as well as in cell lines; and that low-level expression of miR-124 is much lower in highly metastatic ovarian cancer cells and tissues. Meantime, overexpression of miR-124 dramatically inhibits the motility of ovarian cancer cells in vitro and substantially suppresses the protein expression of SphK1, reported as an invasion and metastasis-related gene in human cancers, whose expression is markedly increased in both ovarian cancer cell lines and clinical samples, particularly in two highly metastasis cells, SKOV3-ip and HO8910pm as well as metastatic ovarian tumor tissues. Furthermore, SphK1 is identified as a direct target of miR-124, and knock-down of SphK1 in ovarian cancer cells, SKOV3-ip and HO8910pm, could mimic the inhibition of migration and invasion by miR-124, while re-introduction of SphK1 abrogates the suppression of motility and invasiveness induced by miR-124 in both cell lines. Our studies suggest a protective role of miR-124 in inhibition of migration and invasion in the molecular etiology of ovarian cancer, and a potentially novel application of miR-124 in the regulation of migration and invasion in EOC.
Article
Full-text available
microRNAs (miRNAs/miRs) may have a crucial function in tumor metastasis through the regulation of a plethora of signaling pathways. Increasing evidence has shown that miR-199a is important in regulating the tumor metastasis of ovarian cancer, although the precise biological function of miR-199a is unclear at present. In the current study, it was observed that the expression levels of miR-199a were higher in OV2008 cells compared with C13* cells. However, lower levels of mammalian target of rapamycin (mTOR) protein were detected by western blotting in the OV2008 cells compared with the C13* cells. The miR-199a levels were increased in the C13* cells using miR-199a mimics and the mTOR levels were observed to decrease. This may have resulted in a reversal of cisplatin resistance in the C13* cells. To test this hypothesis, the Renilla luciferase reporter gene system was used to analyze the mTOR levels. The results indicated that the expression levels of mTOR were significantly blocked by the increased miR-199a levels. When the miR-199a inhibitor was applied to decrease the miR-199a levels, it was observed that the mTOR expression levels were increased, while cisplatin-induced apoptosis was decreased in the OV2008 cells. The study concludes that miR-199a is able to reverse cisplatin resistance in human ovarian cancer cells through the inhibition of mTOR and that mTOR may be the target of miR-199a during this process.
Article
Full-text available
Cancer stem cells (CSCs) are believed to be 'seed cell' in cancer recurrence and metastasis. MicroRNAs (miRNAs) can play an important role in the progression of primary tumor towards metastasis by regulating the epithelial-mesenchymal transition (EMT). The goal of this study was to investigate the effect of miRNA-200c overexpression on the EMT, tumorigenicity and metastasis of epithelial ovarian cancer (EOC) CSCs. The EOC CD117+CD44+CSCs were isolated from the human ovarian cancer cell line SKOV3 by using a magnetic-activated cell sorting system, and the lentivirus miR-200c transduced CSCs were then selected for the study. The assays of colony forming, wound healing, cellular migration in vitro and tumor progression in vivo were performed. The miR-200c expression was reduced in the CD117+CD44+CSCs compared with the non-CD117+CD44+CSCs. However, the stable overexpression of the miR-200c in the CD117+CD44+CSCs resulted in a significant down-regulation of ZEB-1 and the Vimentin expression, an upregulation of the E-cadherin expression as well as a decrease of colony forming, migratory and invasion in vitro. Importantly, the miR-200c overexpression significantly inhibited the CD117+CD44+CSCs xenograft growth and lung metastasis in vivo in nude mice by inhibition of the EMT. In addition, the down-regulation of ZEB-1 showed the same efficacy as the miR-200c overexpression in the CD117+CD44+CSCs. These findings from this study suggest that the miR-200c overexpression may be considered a critical approach for the EOC CD117+CD44+CSCs in clinical trials.
Article
Full-text available
The epithelial-to-mesenchymal transition (EMT) that occurs during embryonic development is recapitulated during tumor metastasis. Important regulators of this process include growth factors, transcription factors, and adhesion molecules. New evidence suggests that microRNA (miRNA) activity contributes to metastatic progression and EMT; however, the mechanisms leading to altered miRNA expression during cancer progression remain poorly under- stood. Importantly, overexpression of the epidermal growth factor receptor (EGFR) in ovarian cancer correlates with poor disease outcome and induces EMT in ovarian cancer cells. We report that EGFR signaling leads to transcriptional repression of the miRNA miR-125a through the ETS family transcription factor PEA3. Overexpression of miR-125a inducesconversionofhighlyinvasiveovariancancercellsfromamesenchymaltoanepithelialmorphology,suggesting miR-125a is a negative regulator of EMT. We identify AT-rich interactive domain 3B (ARID3B) as a target of miR-125a and demonstrate that ARID3B is overexpressed in human ovarian cancer. Repression of miR-125a through growth factor signaling represents a novel mechanism for regulating ovarian cancer invasive behavior.
Article
Full-text available
Epithelial ovarian cancer is the most lethal gynecologic malignancy; it is highly aggressive and causes almost 125,000 deaths yearly. Despite advances in detection and cytotoxic therapies, a low percentage of patients with advanced stage disease survive 5 y after the initial diagnosis. The high mortality of this disease is mainly caused by resistance to the available therapies. Here, we profiled microRNA (miR) expression in serous epithelial ovarian carcinomas to assess the possibility of a miR signature associated with chemoresistance. We analyzed tumor samples from 198 patients (86 patients as a training set and 112 patients as a validation set) for human miRs. A signature of 23 miRs associated with chemoresistance was generated by array analysis in the training set. Quantitative RT-PCR in the validation set confirmed that three miRs (miR-484, -642, and -217) were able to predict chemoresistance of these tumors. Additional analysis of miR-484 revealed that the sensitive phenotype is caused by a modulation of tumor vasculature through the regulation of the VEGFB and VEGFR2 pathways. We present compelling evidence that three miRs can classify the response to chemotherapy of ovarian cancer patients in a large multicenter cohort and that one of these three miRs is involved in the control of tumor angiogenesis, indicating an option in the treatment of these patients. Our results suggest, in fact, that blockage of VEGF through the use of an anti-VEGFA antibody may not be sufficient to improve survival in ovarian cancer patients unless VEGFB signaling is also blocked.
Article
Full-text available
Serous epithelial ovarian cancer (EOC) patients often succumb to aggressive metastatic disease, yet little is known about the behavior and genetics of ovarian cancer metastasis. Here, we aim to understand how omental metastases differ from primary tumors and how these differences may influence chemotherapy. We analyzed the miRNA expression profiles of primary EOC tumors and their respective omental metastases from 9 patients using miRNA Taqman qPCR arrays. We find 17 miRNAs with differential expression in omental lesions compared to primary tumors. miR-21, miR-150, and miR-146a have low expression in most primary tumors with significantly increased expression in omental lesions, with concomitant decreased expression of predicted mRNA targets based on mRNA expression. We find that miR-150 and miR-146a mediate spheroid size. Both miR-146a and miR-150 increase the number of residual surviving cells by 2-4 fold when challenged with lethal cisplatin concentrations. These observations suggest that at least two of the miRNAs, miR-146a and miR-150, up-regulated in omental lesions, stimulate survival and increase drug tolerance. Our observations suggest that cancer cells in omental tumors express key miRNAs differently than primary tumors, and that at least some of these microRNAs may be critical regulators of the emergence of drug resistant disease.
Article
Evidence has indicated that ovarian epithelial cancer-type cells under serum-free culture conditions can form spheroid cells and exhibit characteristics expected of cancer stem-like cells (CSCs). However, the mechanism by which differentiated ovarian cancer cells acquire stem-cell properties during CSC enrichment has needed to be elucidated. Recent studies have demonstrated that induction of epithelial to mesenchymal transition (EMT) can generate CSCs and be associated with tumour aggressiveness and metastasis. Ovarian epithelial cancer cell lines, SKOV3 and HO8920, were cultured for spheroid cells and adherent cells. CSC enrichment was investigated using MTT assay, flow cytometery and qRT-PCR and expression level of PI3K/AKT pathway components was analysed by western blotting. Compared to adherent cells, the spheroid cells expressed mesenchymal markers highly and exhibited significantly more motility; we also observed increases in phosphate AKT1 levels in the spheroid cells. Moreover, transfection of miR-20a or miR-200c led to corresponding reduction in endogenous PTEN protein, while AKT1 and phosphate AKT1 levels were upregulated in miRNAs-transfected cells. Finally, PI3K/AKT pathway inhibitor LY294002 reduced expressions of mesenchymal markers and stem-cell gene activity in spheroid cells, enhancing sensitivity of spheroid cells to paclitaxel treatment. Our findings demonstrate that EMT contributed to enrichment of ovarian CSCs in vitro, making EMT targeting in epithelial ovarian cancer a novel therapeutic option.