Вы находитесь на странице: 1из 14

The Role of MicroRNA miR142, miR302, and miR503 to inhibit Endometrioid-

Endometrial Carcinoma Cells Proliferation by Targeting Cyclin-D1 Pathway

Aiman Hilmi Asaduddin1, Ardhia Fefrine Indarta2, Indriaswari Kirana Suri3

1
Medical Student, Faculty of Medicine, Sebelas Maret University,
2
Medical Student, Faculty of Medicine, Sebelas Maret University,
3
Medical Student, Faculty of Medicine, Sebelas Maret University,

Abstract

Endometrial cancer especially endometrioid-endometrial cancer (EEC) is the most common gynecological cancer
worldwide. The proliferation process of EEC is controlled by protein called Cyclin-D1 (CD1). Certain types of
MicroRNAs (miRNAs), named miR-142, miR-302 and miR-503, are down-regulated in EEC and have potential role to
inhibit EEC cells migration and proliferation and promotes cancer cells apoptosis. We aimed to explore the potency of
these miRNAs as a treatment that blocked cell-cycle. The method used four search engines that continued by 4
inclusions and 1 exclusion criteria to be analyzed. Our review was arranged by 48 Journals selected by this method. The
results evaluated Ishikawa cell injected in nude mice using luciferase assay, xenograft and fluorescent reporter assay.
Besides, this study also reviewed researches that use Western blot and the Annexin V-FITC/PI double-staining assay to
detect the miRNAs ability on EEC proliferation. Present miRNAs had been proved that they have same binding site on
their sequence, C-terminal cyclin-D1 gene (CCND1). These miRNAs could down-regulate the CD1 activity, which
regulate cancer proliferation. Two of them, miR-142 and miR-503, blocked on specific phase that resulted in G0/G1-S
phase arrest, while miR-302 could arrest G0/G1-S and G2-M phases. In addition, miRNAs led to apoptosis of EEC cell
via various pathways. Currently, the treatment options for EEC are limited. Palbociclib, rapamycin, paclitaxel and
metformin are some of treatment that give promising anti-growth for patient. Unfortunately, those treatments have side
effect for long-term used such as increase gastrointestinal toxicity and sporadic lung disease. Compared to those
treatments, miRNAs have potential activity against tumor progression in EEC especially in early stage. They enhanced
rapidly to the subset of anti-growth. In conclusion, miR-142, miR-302 and miR503 could induce anti-growth, reduce the
risk of metastasis and trigger apoptosis. These miRNAs could be potential therapy against EEC in following clinical
application.

Keywords: miR-142, miR-302, miR-503, Cyclin-D1, EEC


1. Introduction kinases (CDKs)12. There is one type of cyclin

Endometrial cancer (EC) is the most which important for regulation of proliferation,

common gynecological cancer as it has differentiation and transcriptional control

become more complex for its histological called Cyclin-D1 (CD1)13.

changes and treatment progress1. The CD1, which encoded by C-terminal cyclin-

incidence has increased by 2.3 % per year and D1 gene (CCDN1) lied on 11q13, adhere to

there is no age-standardized2. The main risk CDK4 or CDK6 to form CD1/CDK4/6

factor is endogenous estrogen that links to complexes14. These complexes allow cells to

nulliparity, early menarche, late menopause, enter early G1 phase. Late G1 phase is

and obesity3,4. Those contribute on the rising activated by phosphorylation of another

of estrogen concentration and increase protein15. It will induce release of transcription

mutagenic activity of endometrial cells5,6. factors and DNA is subsequently being

This cancer can be distinguished by its replicated thus transition of cells from G1 to S

microscopic features. Type I is endometrioid phase could occur16.

endometrial carcinoma (EEC) and type II is Endometrial cells over-proliferation

serous carcinoma7. EEC covers 80%-90% of happened because of accumulation of CD1 by

all EC patients and associated with elevated 2 main factors, growth factors excess or DNA

estrogen and endometrium hyperplasia8. damages. One form of DNA damage is

Although the prognosis of EEC still good, it CCND1 mutation14. CD1 c-terminus contains a

can get worse if the patient did not get proper region that have functions in CD1 multi-steps

treatment. Over 8%-11% EEC patients have of destruction, including phosphorylation,

developed into metastasis stage9. When nuclear export, ubiquitination, and protein

prognosis getting worse, it could be continued degradation. Phosphorylation of CD1 induces

by surgical or therapy management with nuclear export and protein broken down in

complications possibility as the side effect, cytoplasm. Mutation at c-terminus of CD1

such as bladder injury, lymphatic leads to inhibition of CD1 phosphorylation

complication, and bleeding complication10,11. thus multi-steps of degradation, resulting in

The origin of hyperplasia in endometrium is CD1 nuclear accumulation. Furthermore, pile

from the abnormality of cell cycle. This of CD1 nuclear activates CDK4/CDK62.

abnormality took place in critical phase of cell These can lead to dysregulation of

cycle called transition phase from G1 to S, CD1/CDK4/6 complexes and subsequently

whereas DNA replication occurs. This phase is diminish G0 phase mechanism and accelerate

driven by cyclins and cyclin-dependent the cell to enter early G1 phase. The
dysregulation is characterized by subtle but in endometrial serous adenocarcinoma cells
irreversible oncogenic change that can be compared with normal endometrium cells20.
obtained by avoiding oncogene-induced Interestingly, there are 3 miRNAs, namely
apoptosis activation15. miR-142, miR-302, and miR-503, which play
Besides, the development of endometrial important role in cyclin-D1 pathway of
cancer pathogenesis and prognosis studies endometrial cancer progression. Those
showed an interest about the role of miRNAs are down-regulated in endometrial
microRNAs (miRNAs). miRNA is a class of cancer and have potential role to inhibit
small RNAs (size of 20–25 nucleotides) that endometrial cancer cells migration and
play a major role in post-transcriptional proliferation and promotes cancer cells
regulation of genes17. miRNAs regulate apoptosis17. Therefore, the aim of this
several mechanisms in cell cycle including literature review is to determine the role of
developmental timing, stem cell division, miR-142, miR-302, and miR-503 in EEC via
senescence and apoptosis17. Several miRNAs inhibiting regulation of cyclin-D1.
have been associated with the development of
2. Methods
different cancer types18. miRNAs involved in
cancer are classified into oncogenic miRNAs The topic of this literature review is

(oncomiRs), which are up-regulated in tumor determined based on problem analysis of

tissues; and tumor suppressor miRNAs (tumor genital health in Indonesia who hasn't reached

suppressor miRs). Use of miRNAs may the target at Sustainable Development Goals

contribute to determine oncogenic and 2030. This literature review discusses about

homeostatic mechanisms19. Accordingly, three main journals regarding the role of miR-

miRNAs have potential utilization as treatment 142, miR-302, and miR-503 on CCND1, which

through comprehensive regulation of has been systematically sought through search

molecules expression related to cancer19. engine on Google Scholar with 4 inclusion

However, miRNAs are implicated in cancer criteria. Thus, three main journals are obtained.

progression and showed that the expression Other supporting articles are explored

patterns of miRNAs in normal tissues are systematically using various specific keywords

different from cancer tissues. That fact on PubMed, ScienceDirect, Cochrane, and

highlights the potential role of miRNAs in the Google Scholar with 4 inclusions and 1

early diagnosis of cancer20. exclusion criteria. 69 scientific publications are

Vasilaou et al cited that there are 54 down- obtained from this method and subsequently

regulated and 66 up-regulated miRNAs groups these together being synthesized


comprehensively. More detailed search prognosis for long term and inhibits recurrence.
strategy is explained below. The result of CCND1 level (Figure 2B) was
transfected by miR-142 and negative control
(NC) in ISK cell. Compared to (NC), the
transfected cell has low level in CCND121.

Figure 2. CCND1 was a direct target of miR-142.


(A) Schematic of miR-142 binding site in CCND1
3’UTR. (B) CCND1 mRNA expression regulated by
miR-142 in ISK cells was determined using qPCR21

In the other side, in vivo study using six-


week old male athymic nude mice were
inoculated subcutaneous. Using western blot
and ISK cell xenografts method, miR142
Figure 1. Schematic Chart of Literature Review treatment was proved to inhibit the
Method
progression of tumorigenicity in mice based
on tumor volume and weight. In 1 month, the
3. Results
average tumor weight in transfected by miR-
3.1. miR-142
142 is 149 mg while in control group is 300
Previous study in 2018 showed the role of
mg21.
miR-142 suppresses EEC proliferation by
targeting CD1. Some in vitro studies prove that
3.2. miR-302
CCND1 3’UTR is direct target in sample of
Genome-wide gene profiling was required
CD1 by using Ishikawa (ISK) cell (Figure 2A).
to determine the genes expression changes in
The bind of miR-142 and CCND1 3’UTR
Ishikawa cells after miR-302 was high
led to down-regulation of CD1 proliferation
expressed to inhibit the carcinogenesis of
and stimulated apoptosis, which exhibit good
Ishikawa cells. miR-302 may alter thousands
of cellular gene expressions (5561 genes) to expression of ZEB1, suppressed the
reduce the carcinogenicity of Ishikawa cells22. expression of Bcl-2 and promoted the
expression of BAX, which inhibits the
proliferation and migration of Ishikawa and
HEC-1A cells23. In vivo assay results showed
that intra-tumoral injection of Ad-miR-302s
led to significantly higher miR-302
expression and inhibited tumor growth by
60.1% compared with the control group23.
Tumors injected with Ad-miR-302s had
significantly lower CD1 protein expression
relative to tumors injected with Ad-LacZ22.
Figure 3. miR-302 decreased the activity of CD1-
30UTR and the expression of CD1 in the Ishikawa
cells22 3.3. miR-503
miR-503 was reported to bind CCND1,
Based on figure 3, CD1 was down-
down-regulate CD1 expression, inhibiting cell
regulated by miR-302 as cell-cycle checkpoint
proliferation, and subsequently making them
gene. This might give benefits in endometrial
remain at G0/G1 phase23-25. miRNA-503
cancer therapy. Luciferase reporter gene
activity on suppressing endometrial cancer
experiments revealed that miR-302
cells proliferation by in-vitro and in-vivo had
specifically inhibited the activity of CD1 30-
been revealed from previous studies24,25. These
UTR-WT, but had no effect on the activity of
studies showed the decreasing of miR-503 and
CD1 30-UTR-MU22. This finding suggests that
increasing of CCND1 in endometrial cancer
miR-302 directly regulates the expression of
cells compared to normal cells. CCND1 was
the CD1, which arrested cell cycle progression
directly targeted by miR-503 and subsequently
in the G0/G1 phase23.
down-regulated CD1 by inhibit CCND1
In addition, Western blot and the Annexin
expression24. Interaction between miR-503 and
V-FITC/PI double-staining assay were used
CCND1 were examined using fluorescent
to detect the ability of miR-302b-3p/302c-
reporter assay. There were two types of
3p/302d-3p to promote the apoptosis of
reporter vectors including two wild-type
Ishikawa and HEC-1A cells23. Besides, qRT-
reporter vectors contained two binding sites
PCR results showed that overexpression of
and three mutant reporter vectors contained
miR-302 significantly inhibited the
point mutation in first site, second site, and miR-503 to expression of CD1 and
both sites respectively. subsequently EEC tumor growth.

4. Discussion
EEC was preceded by typical
histopathological lesions that designated as
endometrial hyperplasia27. Hyperplasia is
usually associated with exogenous estrogen
stimulation accordingly considered as
endometrial carcinogen. Other general
Figure 3. miR-503 directly and negatively regulates
CCND1 in HEC-1-A and Ishikawa cells24 pathways of EEC carcinogenesis are mutations
in p53 and PTEN tumor suppressor genes27.
The data on figure 4 showed that miR-503
On the other hand, several studies showed
decreases the fluorescent intensity in wild-type
there is overexpression of CD1 in EC. This
and second site mutant reporter vectors of both
finding had been considered to be a promising
HEC-1-A and Ishikawa cells. In contrast, miR-
marker of EEC28-30. Based on previous data,
503 was no longer influences the fluorescent
miR-142, miR-302, and miR-503 had potential
intensity in first or both sites mutant reporter
role on down-regulating CD1 thus
vector24. This implied that miR-503 is directly
subsequently decrease CD1 level in
target CCND1 and subsequently downregulate
endometrial cancer21-25.
CCND1 expression.
In this study, CD1 was explained as a target
In-vivo study was required to confirm all
for miR-142, miR-302, and miR-503. CD1
of the in-vitro results in living creature and
acts as an oncogene in different human
explore the effect of miR-503 on EEC tumor
neoplasia when overexpressed by
cells. HEC-1-A containing miR-503 was
chromosomal translocation, gene amplification
injected to nude cells24. The result showed
and protein stabilization31,32. CD1
that a decrease of EEC tumor containing
overexpression results in CCND1
miR-503 cell in size compared to the control.
rearrangement or amplification31. CD1 is
It also showed the decrease of miR-503 level
activated by upstream inputs, including the
and simultaneously increase of CCND1 level
Wnt-β-catenin and Ras/PI3K signaling
in EEC tumor injected by HEC-1-A cells
pathways31,32. Degradation of CD1 is regulated
24
comparing to the control . This in-vivo study
by GSK3β, a downstream target of the PI3K
confirmed the negative or inhibitory effect of
pathway31,32. The phosphorylation of CD1 at
Thr-286 by GSK3β triggers its nuclear export miR-302 generated the wild type and mutated
and cytoplasmic proteolysis via the 26S cells via CCND1. In the other hand, specific
proteasome31,32. Mutations in the CCND1, binding sequence of miR-503 was discovered
which encoding CD1, could result in by examining two potential binding sites to
oncogenic CD1 activation via inhibition of examine binding site of miR-503 exactly25.
Thr-286 phosphorylation, nuclear export, and This subsequently attached at the first binding
protein degradation, thereby promoting site as the potential target in the EEC cell
accumulation of CD1 nuclear33. Nuclear cycle.
retention of CD1 constitutively activates The in-vivo studies of the miRNAs
CDK4/6 kinases through progression of the exhibited tumorigenicity decrease in miRNAs
G1/S cell cycle phase and thereby promotes treatments. The studies used xenograft method
cellular proliferation and tumorigenesis31,32. in nude mice with different type of cells21,22,25.
CCND1 mutations had frequent co-occurring The studies of miR-142 and miR-302 used
mutations of ARID1A and members of the Ishikawa cells while miR-503 used the HEC-
PI3K signaling pathway, PTEN and PIK3CA, 1-A. The result of miR-142 study revealed that
suggesting that alterations these molecular average miR-142 transfected tumor weight
pathways are also required in addition to was smaller compared to control tumor within
oncogenic nuclear CD1 activation in a subset a month21. The immunohistochemistry studies
of endometrial cancers32. Phosphorylation- also discovered that there was lower Ki67+
dependent nuclear export of CD1 at Thr-286 is than CD1 in miR-142 transfected cells.
suggested to be critical for prevention of Besides, another study found that miR-302
aberrant cell proliferation in vitro32. remarkably inhibited tumor growth by 60.1%
Basically, this pathway led the miRNAs compared to the control tumor cells within 17
bind to 3’ UTR (Untranslated Region) of days22. The miR-503 study also suggested
CCND1 mRNA, which differ in specific suppressive effect of miR-503 on EEC cell-
binding site. The studies were done using derived xenografts25.
luciferase reporter assay21,22,25 and showed Those studies showed that miR-142 could
that ISK wild type cells transfected by three suppress EEC proliferation by down-
types of miRNA had a low luciferase regulating CCND1 in vivo. CCND1 played as
fluorescence intensity compared to the a modulator of CDK4/6 and subsequently
mutated. miR-142 binding site on CCND1 regulated G1/S transition21. miR-302 reduced
mRNA was found by generating the wild type CD1 expression in a post-transcriptional
and binding site of mutated cells21. Besides, phase, thus affecting cell proliferation and cell
cycle progression by arresting the G0/G1-S treatment targeting cell cycle lead to potential
phase23,33 and G2/M phase22. It resulted in agents against EEC. Palbociclib, a CDK4/6
significant G2/M arrest and promoted specific inhibitor, that knockout CD1 had
endometrial cancer cells apoptosis by reduce therapeutic potential against EEC cell lines
the expression of ZEB1, inhibit the expression and xenografts expressing the retinoblastoma
of Bcl-2, and promote the expression BAX in protein (Rb)38,39. Rapamycin and its derivates,
EC23. Conceivably, miR-302 could be used to clearly act as cytostatic agents by arresting
silence the expression of several key genes in cells in the G1 phase. Paclitaxel
cell cycle, global demethylation, apoptosis, simultaneously decrease the activity of
and DNA repair along with BMI-1, CXCR4, p70S6K which is a mitogen‐activated
AKT1/2, Runx-1, and EGFR genes serine/threonine kinase that plays a crucial role
simultaneously34. In the other side, decreased in cell growth40. But this treatment has many
miR-503 level in EEC cells resulted in an side effects such as increase the risk of
increase in CCND1, which contributed to sporadic lung disease, tuberous sclerosis
malignant cellular phenotypes both in vitro complex, neurological disease and imbalance
and in vivo25. miR-503 induced G1 arrest by metabolism because of its role to inhibit
targeting an overlapping set of cell-cycle mTOR signaling pathway41.
35
regulators . miR-503 also promoted cell- Other studies revealed that metformin
cycle arrest through cdc25A degradation36. showed increased expression of p21, a cell-
Those mechanisms led to the blockage of the cylce blocker, which led to cell-cycle arrest in
G1/S transition, which suppressed G1 and G2 phase42. Metformin blocked
proliferation and colony formation activities serum-induced proliferation entry to S phase,
of EEC cell lines25. Some differential expres- resulting in G1 cell cycle arrest. Moreover,
sion genes were discovered and the mRNA metformin induced apoptosis at high doses of
level of p21 and CDK4 were down-regulated treatment by increased caspase-3 activity,
in miR-503-3p overexpression cells, which triggers epithelial apoptosis43. However
indicating that miR-503-3p inhibiting cell metformin affect high-prevalence of vitamine
proliferation is partially mediated by these two B12 deficiency44. Besides, Eupatilin treatment
proteins37. inhibited cell progression through the
Currently, the treatment options for induction of G2/M cell cycle arrest45. Long
advanced endometrial cancer patients are term of Eupatilin treatment induced lower
limited. There were some evidences showed motility of colon and ileum as side effects46.
that inhibition of CD1 expression and Meanwhile, miRNAs treatment turn off
growth and induced apoptosis depend on the DNA repair during progression of EEC. Thus,
type. miRNAs also enhanced very rapidly to a miR-142, miR-302, and miR503 could be
subset of anti-growth47. In addition, miRNA used as potential therapy against EEC in
led to epithelial-to-mesenchymal transition clinical application.
(EMT) process which stop metastasis and
induced apoptosis epithelial cell48. Compared
to other treatments, miRNAs also have
potential activity against tumor progression in
EEC.

5. Conclusion

EEC is the common type of endometrial


cancer worldwide. The origin of EEC occurs
with hyperplasia in endometrium that comes
from cell cycle abnormality. Cyclin-D1 plays
an important role in cell proliferation during
the progression of EEC. Certain type of
miRNAs named miR-142, miR-302, and
miR503 are down-regulated in EEC. These
kind of miRNAs were thought to have
significant role in cell growth thus might be
potential agent against EEC.
Based on this literature review study,
miR-142, miR-302, and miR503 could bind
CCND1 mRNA that cause down-regulated
activity of CD1. Based on in vitro and in vivo
studies, these miRNAs suppressed EEC cells
proliferation and inhibited tumor growth. Two
of them, miR-142 and miR-503, blocked on
specific phase that resulted in G0/G1-S phase
arrest, while miR-302 could arrest G0/G1-S
and G2-M phases. Besides, these miRNAs
also could promote apoptosis and lead to
References 7. Byers T, Sedjo RL. Body fatness as a

1. Philippe Morice, Alexandra Leary, cause of cancer: epidemiologic clues

Carien Creutzberg, Nadeem Abu- to biologic mechanisms. Endocr Relat

Rustum ED, Endometrial. Cancer. 2015;22(3):R125–34.

Endometrial cancer Philippe. Lancet. 8. Dossus L, Allen N, Kaaks R, Bakken

2016; K, Lund E, Tjonneland A, et al.

2. Felix AS, Yang HP, Bell DW, Reproductive risk factors and

Sherman ME. Epidemiology of endometrial cancer: The European

endometrial carcinoma: Etiologic prospective investigation into cancer

importance of hormonal and and nutrition. Int J Cancer. 2010;

metabolic influences. In: Advances in 9. Qie S, Diehl JA. Cyclin D1, cancer

Experimental Medicine and Biology. progression, and opportunities in

2017. cancer treatment. Journal of

3. Lax SF. Pathology of endometrial Molecular Medicine. 2016.

carcinoma. In: Advances in 10. Ghosh A, Kumar M, Shevra C. Cyclin

Experimental Medicine and Biology. D1 and Ki-67 expression in normal,

2017. hyperplastic and neoplastic

4. Press D. The influence of vascular endometrium. J Postgrad Med. 2014;

endothelial growth factor-A and 11. Church DN, Stelloo E, Nout RA,

matrix metalloproteinase-2 and -9 in Valtcheva N, Depreeuw J, Haar N

angiogenesis , metastasis , and Ter, et al. Prognostic significance of

prognosis of endometrial cancer. POLE proofreading mutations in

2017; endometrial cancer. J Natl Cancer

5. Setiawan VW, Yang HP, Pike MC, Inst. 2015;

McCann SE, Yu H, Xiang YB, et al. 12. Geppert B, Persson J. Robotic

Type i and II endometrial cancers: infrarenal paraaortic and pelvic nodal

Have they different risk factors? J staging for endometrial cancer:

Clin Oncol. 2013; Feasibility and lymphatic

6. Felix AS, Weissfeld JL, Stone RA, complications. Acta Obstet Gynecol

Bowser R, Chivukula M, Edwards Scand. 2015;

RP, et al. Factors associated with 13. Mäenpää MM, Nieminen K, Tomás

Type i and Type II endometrial EI, Laurila M, Luukkaala TH,

cancer. Cancer Causes Control. 2010; Mäenpää JU. Robotic-assisted vs


traditional laparoscopic surgery for 271–282. doi:10.1007/s00404-015-
endometrial cancer: a randomized 3660-y
controlled trial. Am J Obstet Gynecol. 18. Olivieri F, Rippo MR, Monsurro` V,
2016; Salvioli S, Capri M, Procopio AD
14. Xu J, Lin DI. Oncogenic c-terminal (2013) MicroRNAs linking inflamm-
cyclin D1 (CCND1) mutations are aging, cellular senescence and cancer.
enriched in endometrioid endometrial Ageing Res Rev 12:1056–1068
adenocarcinomas. Ahmad A, editor. 19. Yanokura M, Banno K, Iida M, Irie
PLoS One [Internet]. 2018 Jul H, Umene K, Masuda K, et al.
3;13(7):e0199688. Available from: Review article : MicroRNAS in
https://dx.plos.org/10.1371/journal.po endometrial cancer : recent advances
ne.0199688 and potential clinical applications.
EXCLI Journal 2015;14:190-198.
15. Narasimha AM, Kaulich M, Shapiro http://dx.doi.org/10.17179/excli2014-
GS, Choi YJ, Sicinski P, Dowdy SF. 590
Cyclin D activates the Rb tumor 20. Vasilatou, D., Sioulas, V. D., Pappa,
suppressor by mono-phosphorylation. V., Papageorgiou, S. G., & Vlahos, N.
Elife. 2014;2014(3):1–21. F. (2015). The role of miRNAs in
16. John R, Malathi N, Ravindran C, endometrial cancer. Epigenomics,
Anandan S. Mini review: 7(6), 951–959. doi:10.2217/epi.15.41
Multifaceted role played by cyclin D1 21. Su Y, Wang J, Ma Z, Gong W, Yu L.
in tumor behavior. Indian J Dent Res miR-142 Suppresses Endometrial
[Internet]. 2017;28(2):187. Available Cancer Proliferation In Vitro and In
from: Vivo by Targeting Cyclin D1 . DNA
http://www.ijdr.in/text.asp?2017/28/2 Cell Biol. 2018;38(2):144–50.
/187/207799 22. Yan G, Yu F, Wang B, Zhou H, Ge
17. Sianou, A., Galyfos, G., Moragianni, Q, Su J, et al. MicroRNA miR-302
D., Andromidas, P., Kaparos, G., inhibits the tumorigenicity of
Baka, S., & Kouskouni, E. (2015). endometrial cancer cells by
The role of microRNAs in the suppression of Cyclin D1 and CDK1.
pathogenesis of endometrial cancer: a Cancer Lett. 2014;345:39–47.
systematic review. Archives of 23. Li, Yibing; Huo, Jianing; Pan, Xin;
Gynecology and Obstetrics, 292(2), Wang, Cuicui; Mi X. MicroRNA
302b-3p/302c-3p/302d-3p inhibits marker for endometrial
epithelial – mesenchymal transition diseases. Diagn Pathol. 2013;8:138.
and promotes apoptosis in human Published 2013 Aug 15.
endometrial carcinoma cells. Onco doi:10.1186/1746-1596-8-138
Targets Ther. 2018;11:1275–84. 29. Yan R, Wu X, Wang Y, Yu C, Li H,
24. Jiang Q, Feng MG, Mo YY. Zhang L. Overexpression of peptidyl-
Systematic validation of predicted prolyl isomerase 1 (Pin1) and cyclin
microRNAs for cyclin D1. BMC D1 in endometrial cancer. Int J Clin
Cancer. 2009;9:3–6. Exp Pathol. 2017;10(3):3335–43.
25. Xu YY, Wu HJ, Ma H Da, Xu LP, 30. Khabaz MN, Ali A, Butt NS, Al-
Huo Y, Yin LR. MicroRNA-503 maghrabi BJ. Cyclin D1 is
suppresses proliferation and cell- significantly associated with stage of
cycle progression of endometrioid tumor and predicts poor survival in
endometrial cancer by negatively endometrial carcinoma patients. Ann
regulating cyclin D1. FEBS J. Diagn Pathol. 2017;30:47–51.
2013;280(16):3768–79. 31. Xu J, Lin DI. Oncogenic c-terminal
26. Hirakawa T, Nasu K, Abe W, Aoyagi cyclin D1 (CCND1) mutations are
Y, Okamoto M, Kai K, et al. MiR- enriched in endometrioid endometrial
503, a microRNA epigenetically adenocarcinomas. PLoS ONE.
repressed in endometriosis, induces 2018;13(7): e0199688.
apoptosis and cell-cycle arrest and https://doi.org/10.1371/journal.pone.0
inhibits cell proliferation, 199688
angiogenesis, and contractility of 32. Ikeda Y, Oda K, Hiraike-wada O,
human ovarian endometriotic stromal Koso T. Cyclin D1 harboring the
cells. Hum Reprod. T286I mutation promotes oncogenic
2016;31(11):2587–97. activation in endometrial cancer.
27. Shevra CR, Ghosh A, Kumar M. Oncol Rep. 2013;30:584–8.
Cyclin D1 and Ki-67 expression in 33. Gao Z, Zhu X, Dou Y. The miR-302 /
normal, hyperplastic and neoplastic 367 cluster : a comprehensive update
endometrium. J Postgrad Med. on its evolution and functions. Open
2019;61(1):15–20. biol. 2015;5:150138.
28. Liang S, Mu K, Wang Y, et al. 34. Yen E, Chen Y, Chen JS, Ying S. The
CyclinD1, a prominent prognostic microRNA and the perspectives of
miR-302. Heliyon [Internet]. kinase 4/6 inhibitor in endometrial
2019;5:e01167. Available from: cancer. PLoS ONE. 2017;12(5):
https://doi.org/10.1016/j.heliyon.2019. e0177019.
e01167 https://doi.org/10.1371/journal.pone.0
35. Forrest AR, Kanamori-Katayama M, 177019
Tomaru Y, Lassmann T, Ninomiya N, 40. Shafer A, Zhou C, Gehrig PA,
Takahashi Y, de Hoon MJ, Kubosaki Boggess JF, Bae-Jump VL.
A, Kaiho A, Suzuki M et al. Induction Rapamycin potentiates the effects of
of microRNAs, mir-155, mir-222, paclitaxel in endometrial cancer cells
mir-424 and mir-503, promotes through inhibition of cell proliferation
monocytic differentiation through and induction of apoptosis. Int J
combinatorial regulation. Leukemia. Cancer. 2010;126(5):1144–1154.
2010;24:460–466. doi:10.1002/ijc.24837
36. Sarkar S, Dey BK & Dutta A. MiR- 41. Li J, Kim SG, Blenis J. Rapamycin:
322/424 and-503 are induced during one drug, many effects. Cell Metab.
muscle differentiation and promote 2014;19(3):373–379.
cell cycle quiescence and doi:10.1016/j.cmet.2014.01.001
differentiation by down-regulation of 42. Takahashi A, Kimura F, Yamanaka A,
Cdc25A. Mol Biol Cell. 2010;21: et al. Metformin impairs growth of
2138–2149. endometrial cancer cells via cell cycle
37. Ren Z, Xue G, Chen Y, Du Y, Pan D, arrest and concomitant autophagy and
Li X, et al. miR-503-3p suppresses apoptosis. Cancer Cell Int.
cell viability and promotes cell 2014;14:53. Published 2014 Jun 16.
apoptosis in cancer cells. Int J Clin doi:10.1186/1475-2867-14-53
Exp Pathol. 2016;9(11):10943–54. 43. Cantrell LA, Zhou C, Mendivil A,
38. Dosil MA, Mirantes C, Eritja N, Felip Malloy KM, Gehrig PA, Bae-Jump
I, Navaridas R, Gatius S, et al. VL. Metformin is a potent inhibitor of
Palbociclib has antitumour effects on endometrial cancer cell proliferation--
Pten-deficient endometrial neoplasias. implications for a novel treatment
J Pathol. 2017;242(2):152-164. strategy. Gynecol Oncol.
39. Tanaka T, Terai Y, Ashihara K, 2010;116(1):92–98.
Fujiwara S, Tanaka Y, Sasaki H, et al. doi:10.1016/j.ygyno.2009.09.024
The efficacy of the cyclin-dependent
44. Kos E, Liszek M, Emanuele M, 2014;6:205–216. Published 2014 Apr
Durazo-Arvizu R, Camacho P. Effect 25. doi:10.2147/CMAR.S38156
of Metformin Therapy on Vitamin D
and Vitamin B 12 Levels in Patients
with Type 2 Diabetes Mellitus.
Endocr Pract. 2012; 18(2):179-84.
doi: 10.4158/EP11009.OR.
45. Cho JH, Lee JG, Yang YI, Kim JH,
Ahn JH, Baek NI, et al. Eupatilin, a
dietary flavonoid, induces G2/M cell
cycle arrest in human endometrial
cancer cells. Food Chem Toxicol.
2011; 49(8):1737-44. doi:
10.1016/j.fct.2011.04.019.
46. Ryoo SB, Oh HK, Yu SA, et al. The
effects of eupatilin (stillen®) on
motility of human lower
gastrointestinal tracts. Korean J
Physiol Pharmacol. 2014;18(5):383–
390. doi:10.4196/kjpp.2014.18.5.383
47. Brachova P, Mueting SR, Devor EJ,
Leslie KK. Oncomorphic TP53
Mutations in Gynecologic Cancers
Lose the Normal Protein:Protein
Interactions with the microRNA
Microprocessing Complex. J Cancer
Ther. 2014;5(6):506–516.
doi:10.4236/jct.2014.56058
48. Díaz-López A, Moreno-Bueno G,
Cano A. Role of microRNA in
epithelial to mesenchymal transition
and metastasis and clinical
perspectives. Cancer Manag Res.

Вам также может понравиться