Mutually distinguishing microRNA signatures of breast, ovarian and endometrial cancers in vitro

  • Authors:
    • Marc Hirschfeld
    • Isabel Ge
    • Gerta Rücker
    • Julia Waldschmidt
    • Sebastian Mayer
    • Markus Jäger
    • Matthias Voigt
    • Bernd Kammerer
    • Claudia Nöthling
    • Kai Berner
    • Daniela Weiss
    • Jasmin Asberger
    • Thalia Erbes
  • View Affiliations

  • Published online on: August 27, 2020     https://doi.org/10.3892/mmr.2020.11466
  • Pages: 4048-4060
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Early diagnosis and therapy in the first stages of a malignant disease is the most crucial factor for successful cancer treatment and recovery. Currently, there is a high demand for novel diagnostic tools that indicate neoplasms in the first or pre‑malignant stages. MicroRNAs (miRNA or miR) are small non‑coding RNAs that may act as oncogenes and downregulate tumor‑suppressor genes. The detection and mutual discrimination of the three common female malignant neoplasia types breast (BC), ovarian (OC) and endometrial cancer (EC) could be enabled by identification of tumor entity‑specific miRNA expression differences. In the present study, the relative expression levels of 25 BC, EC and OC‑related miRNAs were assessed by reverse transcription‑quantitative PCR and determined using the 2‑ΔΔCq method for normalization against the mean of four housekeeping genes. Expression levels of all miRNAs were analyzed by regression against cell line as a factor. An expression level‑based discrimination between BC and OC cell types was obtained for a subgroup of ten different miRNA types. miR‑30 family genes, as well as three other miRNAs, were found to be uniformly upregulated in OC cells compared with BC cells. BC and EC cells could be distinguished by the expression profiles of six specific miRNAs. In addition, four miRNAs were differentially expressed between EC and OC cells. In conclusion, miRNAs were identified as a potential novel tool to detect and mutually discriminate between BC, OC and EC. Based on a subset of 25 clinically relevant human miRNA types, the present study could significantly discriminate between these three female cancer types by means of their expression levels. For further verification and validation of miRNA‑based biomarker expression signatures that enable valuable tumor detection and characterization in routine screening or potential therapy monitoring, additional and extended in vitro analyses, followed by translational studies utilizing patients' tissue and liquid biopsy materials, are required.

Introduction

The success rate in the clinical treatment of neoplastic disease remains highly associated with early detection of pre-malignant or first stages of malignant tissues. To date, only few highly specific and sensitive biomarkers are routinely used in the clinic for early-stage cancer screening or diagnostics.

Due to mammography screening, which was first introduced in 2005 in Germany, breast cancer (BC) has been identified at earlier stages, when treatment options are most promising and prognosis is most favorable (1). For endometrial cancer (EC) and ovarian cancer (OC), no standardized screening has yet been established. Postmenopausal bleeding serves as an early indicator of EC (2,3) European studies have shown that the 5-year survival rate of endometrial adenocarcinoma is >90% when detected at stage I compared with a survival rate of ~50% for advanced stages (II, III, IV) (3,4). OC remains one of the most challenging types of cancer to detect and treat. In most cases, tumor progression and metastasis are unnoticed until the advanced stages (5). According to the Surveillance, Epidemiology and End Results Program (National Cancer Institute, USA) database, the 5-year survival rate for localized disease is >90% in the USA population (6), however only 20% of ovarian cancer cases are detected at such an early stage in the USA (5,7).

One possible approach in the identification of novel potential biomarker candidates is based on expression profiling of different states, for example comparing malignant and healthy control expression profiles (7). In a stepwise filtering process, the discovery, qualification, verification, potential candidate prioritization and subsequent validation in adequate cohort sizes demonstrate the applicability of a biomarker for clinical practice implementation (7). Among a multitude of potential biomarker types, in previous years one group of nucleic acids has gained significant attention due to their diverse regulatory functions (8).

MicroRNAs (miRNAs or miRs) are small non-coding RNA molecules of ~22 nucleotides in length, which are involved in the post-transcriptional regulation of gene expression, predominately via gene silencing. By binding to various mRNA targets, upregulation of miRNA leads to reduced translation of mRNA or degradation of its transcript (9). In cancer, dysregulated miRNA expression plays an important role by upregulating oncogenes and downregulating tumor-suppressor genes, thus modulating cell proliferation, differentiation, apoptosis and stress response (10). The regulatory influence of miRNAs in breast and gynecological cancer biology has been demonstrated in a growing number of studies (8,1117). The selection of miRNAs in the present study was based on an extensive literature search, with the major criterion being expression changes in the tumor types of BC, EC and OC (Table I), in combination with a proven detectability of all analyzed miRNA types in in vitro models as well as in human urine samples (1820).

Table I.

miRNA types with functional implications in breast, endometrial and ovarian cancers.

Table I.

miRNA types with functional implications in breast, endometrial and ovarian cancers.

miRNATarget genesBreast cancerEndometrial cancerOvarian cancer(Refs.)
Let-7aCCL21, CCR7, RAS, HMGA-11, HMGA-2, cyclin A2,Plasma ↑ (53)Tissue ↓ (58)Cell line ↓ (59)(28,5661)
CDC34, STK6, STK12, E2F5, CDK8, CDC25A, CDK6,Tissue ↓ (54,55)
Casp3, Bcl2, Map3k1, Cdk5Cell line ↓ (5456)
Tissue ↓ (57)
Let-7b Tissue ↓ (57)Cell line ↓ (60)Tissue ↓ (61)(28,60,6264)
Serum ↓ (28)
Cell line ↓ (59)
Let-7c Tissue (endometrioid) ↑ (62)Cell line ↓ (59)(28,65,66)
Tissue (Sarcoma, mixed epithelial-
mesenchymal tumors) ↓ (63)
miR-21ANP32A, BTG2, Bcl2, P12/CDK2AP1, HNRPK,Tissue ↑ ((53,64)Cell line ↑ (22)Cell line ↑ (66)(19,56,6769)
IL-12p35, JAG1, MEF2C, hMSH2, PDCD4, PTEN,CTCs ↑ (64)
RECK, RhoB, SMARCA4, TGFBRIIPlasma ↑ (64)
Cell lines ↑ (65)
miR-27aWnt, PPARγ, C/EBPα, FOXO1Tissue ↓ (65)Tissue (67)Cell lines ↑ (46)(46,68,70)
Cell lines ↑ (67)
miR-30aITGB3, UBC9, TP53, TRADD, CCNE1 (NF-κB)Cell lines ↓ (65,68,69)Cell line ↓ (60)Cell line (Clear Cell) ↑ (72)(57,68,71,76)
Tissue → (70)Tissue ↓ (71)Tissue (Clear cell) ↑ (73)
Plasma ↓ (71)Urine ↑ (74) Plasma ↓ (75)
miR-30c Tissue ↓ (77)Tissue ↓ (62,78)Tissue ↑ (34)(31,65,7982)
Whole blood ↑ (76)
Cell line (drug-resistant) ↓ (79)
miR-30e Plasma ↓ (53)Tissue (papillary serous vs.Tissue ↑ (35)(31,56,82)
Tissue ↓ (53)Endometrioid) ↑ (79)
miR-100mTOR, PLK-1, FRAK1, Wnt/β-CateninCell lines ↓ (80)Tissue ↓ (38)Tissue ↓ (38)(35,82,83)
Tissue ↓ (38) Serum ↓ (81)
miR-7EGFR, IGF1R, PIK3CD, KLF4CSCs ↓ (17,82)Tissue ↑ (17)Tissue ↑ (83)(17,37,84)
miR-125bERBB2, ERBB3, BCL3, EPORTissue ↓ (65)Cell line ↓ (37)Tissue ↓ (83)(68,83,81,84)
Serum ↓ (81)
miR-9REST, CoRESTCell lines ↑ (65)Tissue ↑ (67)Tissue ↑ (83)(15,59,75,84)
Cell lines ↑ (67)
Plasma ↓ (15)
miR-15bBCL2, CHEK1Cell lines ↓ (65)Tissue ↓ (84)Tissue ↓ (83)(68,84,85)
miR-128.1R3HDM1, RCSCell lines ↓ (65)Tissue ↑ (67)Cell lines ↓ (81)(68,75,86)
Cell lines ↑ (67)
miR-222CD117, PBX3Cell lines ↑ (65)Serum ↑ (17)Tissue ↓ (83)(17,68,84)
Serum ↑ (65)
miR-29STAT3, MCL1, TCL1A, TTP, DNMT3Cell lines ↑Tissue ↓ (84)Serum ↑ (85)(68,85,87)
Serum ↑ (65)
miR-92aERβ, MUC16Cell lines ↓ (65)Tissue ↑ (15)Serum ↑ (85)(15,68,87)
Plasma ↑ (15)
miR-200ZEB1, ZEB2, β tubulin IIICell lines ↓ (65)Cell lines ↑ (17,86)Tissue ↑ (83)(15,17,60,68,8385,88)
Tissue ↑ (57)Tissue ↑ (15,84)Serum ↑ (81)
Plasma ↑ (15)
miR-17NOR-1, GALNT3Cell lines ↓ (87)Cell lines ↓↑ (86) (88,89)
miR-20NOR-1, PTEN/PI3K/AktCell lines ↓ (87)Cell lines ↑ (86)Tissue ↓ (83)(84,85,88,89)
Tissue ↓ (84)
miR-19bMfn1, PITX1, ATXN1, PTEN, Sbf2, Bcl7a, Rnf44Cell lines ↓ (87)Cell lines ↓ (86)Tissue ↑ (88)(8890)
miR-106bE2F5, SLC2A3, E2F1, TWST1Cell lines ↓ (87)Tissue ↑ (17)Tissue/Cell line (89)   (17,89,91)
miR-221CD117, SND1, AEG-1Cell lines ↑ (65)Tissue ↓ (17, 84)Serum ↑ (85)(17,68,85,87,91)
Serum ↑ (65) Tissue/Cell line (89)

[i] ↑, upregulated expression; ↓, downregulated expression; miR/miRNA, microRNA.

miRNA-21 (miR-21) is one of the most common miRNAs in epithelial cancer, and it generally promotes anti-apoptotic effects in various malignant tissues and cell lines, including BC, OC and EC, by downregulating tumor suppressors, such as phosphatase and tensin homolog (21,22) and programmed cell death protein 4 (23). In patients with BC, overexpression of miR-21 in the tumor is associated with advanced tumor stage, lymph node metastasis and poor survival (24). Whereas, in OC cell lines, miR-21 promotes pathways that enhance chemoresistance (25).

In contrast to miR-21, members of the miRNA family let-7 have most commonly been reported as tumor suppressors by downregulating Harvey rat sarcoma viral oncogene homolog and high-mobility group AT-hook 2 (26). However, studies have reported inconsistent results regarding the individual member let-7b. While some studies reported that high levels of let-7b in serum and plasma was associated with a favorable prognosis in cancer (27,28), a previous meta-analysis demonstrated reduced survival rates in high-grade serous OC with high tissue expression of let-7b (29). The tumor suppressing miRNA family miR-30 has been reported to exhibit pro-apoptotic effects by silencing ubiquitin-conjugating enzyme 9 and integrin β3 (30). In BC, miR-30a inhibits cell migration and invasion (31), whereas expression of miR-30c in tissues is associated with benefits during endocrine treatment (32) and regulatory effects in chemotherapy resistance processes (33). Notably, high expression levels of miR-30c and miR-30e have been observed in OC compared with normal tissue; however, both miRNAs are associated with an improved prognosis (3436).

A more homogenous profiling has been observed for miR-125b and miR-100. miR-125b and miR-100 mediate the Erb-B2 receptor tyrosine kinase 2 and mechanistic target of rapamycin pathways, respectively, and downregulation of both miRNAs has been reported in BC, OC and EC tissue and cell lines (3741). The previously described functional implications of the investigated miRNAs in BC, EC and OC tumor biology are summarized in Table I.

Due to recent investigations on miRNAs that are commonly conducted based on different study designs and environments, the comparison and interpretation of results between multiple cancer types have become increasingly challenging. The goal of the present study was to evaluate differences of miRNA profiling in three of the most common female cancer types: BC, OC and EC. Instead of solely focusing on individual miRNA types or families, the present study aimed to investigate the expression patterns of miRNAs that have great potential to serve as promising diagnostic tools in the distinction of different tumor types. Based on three cell types for each type of malignancy, BC, OC and EC, the detected differences in quantitative expression levels of a set of 25 miRNAs revealed diagnostic biomarker features clustered in tumor-entity-specific ‘miRNA signatures’. To this end, the in vitro models used were selected to represent a range of common subtypes/properties of the respective carcinomas. The data obtained in this first phase biomarker identification study serve as a basis to prioritize distinct miRNAs with diagnostic significance that will be investigated in future studies.

Materials and methods

Cell culture conditions and treatments

The BC cell lines BT-20 (cat. no. 300130; CLS Cell Lines Service GmbH), BT-474 (cat. no. 00131; CLS Cell Lines Service GmbH) and SK-BR-3 (cat. no. 300333; CLS Cell Lines Service GmbH), the EC cell lines Ishikawa (cat. no. 99040201; Sigma-Aldrich; Merck KGaA), EFE-184 (cat. no. ACC 230; Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures GmbH) and AN3CA (cat. no. 300119; CLS Cell Lines Service GmbH), and the OC cell lines SK-OV-3 (cat. no. 300342; CLS Cell Lines Service GmbH), EFO-27 (cat. no. ACC 191; Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures GmbH) and OAW-42 (cat. no. 300304; CLS Cell Lines Service GmbH) were incubated in a humidified atmosphere at 37°C and 5% CO2. Ishikawa cells were cultured in RPMI-1640 (Gibco; Thermo Fisher Scientific, Inc.) supplemented with 10% newborn calf serum (Gibco; Thermo Fisher Scientific, Inc.), 1% HEPES buffer (Gibco; Thermo Fisher Scientific, Inc.) and 100 U/ml Penicillin/Streptomycin (Sigma-Aldrich; Merck KGaA). The BT-20, SK-BR-2, EFE-184, AN3CA, SK-OV-3 and EFO-28 cells were cultured in DMEM/F12 (cat. no. 31331-028; Thermo Fisher Scientific, Inc.) supplemented with 10% newborn calf serum, 1% HEPES buffer and 100 U/ml Penicillin/Streptomycin. The BT-47 and OAW-42 cells were cultured in DMEM/F12 supplemented with 2.5% insulin (Insuman rapid®; Sanofi S.A.).

miRNA isolation

miRNA from cultured cells was isolated using the innuPREP Micro RNA kit (Analytik Jena US LLC), according to the manufacturer's instructions. Isolated RNA was quantitatively determined using the NanoDrop ND1000 (VWR International GmbH). RNA samples were stored at −20°C until further processing.

Reverse transcription-quantitative PCR (RT-qPCR)

Total RNA was isolated using GeneMATRIX Universal RNA/miRNA Purification kit (cat. no. E3599; EURx®; Roboklon GmbH) according to manufacturer's protocol. A total of 1 µg isolated RNA per sample was used for RT. The RT reaction mix contained 5 µl RT-buffer (5X), 1 µl 2.5 µM poly A adapter primer (Apara Bioscience GmbH), 0.5 µl 5 mM dNTPs (Jena Bioscience), 0.25 µl Maxima reverse transcriptase (Thermo Fisher Scientific, Inc.), 0.25 µl SUPERase In RNase inhibitor (Thermo Fisher Scientific, Inc.), 0.5 µl 10 mM ATP (New England Biolabs, Inc.), 0.25 µl poly A polymerase, and 1 µg RNA sample. The reaction was performed on a thermal cycler (Eppendorf) at 37°C for 60 min and stopped at 85°C for 10 min. Processed cDNA was stored at 4°C.

The relative expression levels of specific miRNAs were assessed by qPCR using the SYBR-Green assay in a duplicate analysis. A total of 1 µl cDNA per sample with a concentration of 5 ng/µl was mixed with 9 µl Master Mix, containing 1 µl buffer (10X), 0.5 µl 5 mM dNTPs (Jena Bioscience GmbH), 0.5 µl 5 µM primer (Apara), 0.5 µl SYBR-Green (Roche Diagnostics), 0.05 µl HotStart Taq (Jena Bioscience GmbH) and 6.45 µl nuclease-free water (Analytik Jena US LLC). The primer pairs consisted of a universal reverse primer (3032) and a specific miRNA sense primer. The qPCR was performed on a LightCycler® 480 instrument (Roche Diagnostics) at 95°C for 5 min, followed by 40 cycles at 95°C for 5 sec, 62°C for 15 sec and 72°C for 10 sec. Data were analyzed with the LightCycler® 480 software (Roche Molecular Systems, Inc.; Version 1.5.1). The relative expression of each miRNA was determined using the 2−ΔΔCq method (42,43) based on the housekeeping genes small nucleolar RNA, C/D box 48 (RNU48), miR-26b, miR-16 and miR-103, with the ‘BestKeeper’ software tool (Version 1) (43). The specific primer sequences are listed in Table II.

Table II.

Primer sequences.

Table II.

Primer sequences.

PrimerSequence (5′→3′)
miRNA poly GAAGACTCAGTTGCACTCTACCAAT
A RT TAAGACGAACAGAGCCATACTTTTTTTTTTTTNN
Universal antisense GACTCAGTTGCACTCTACCAATTAA
miR-16-5p GGCTAGCAGCACGTAAATATTG
miR-26b-5p GGCGTTCAAGTAATTCAGGATAG
RNU48 TGTGTCGCTGATGCCATC
miR-103-5p CGGAGCAGCATTGTACAGG
let-7a-5p CGGTGAGGTAGTAGGTTGTATAGTT
let-7b-5p CGTGAGGTAGTAGGTTGTGTG
let-7d-5p CGGAGAGGTAGTAGGTTGCATA
miR-7-5p CGGTGGAAGACTAGTGATTTTGTT
miR-9-5p CGGTCTTTGGTTATCTAGCTGTAT
miR-15b-5p GCTAGCAGCACATCATGGTTTA
miR-17-5p GCAAAGTGCTTACAGTGCAG
miR-19b-3p GTGTGCAAATCCATGCAAAACT
miR-20a-5p CGGTAAAGTGCTTATAGTGCAGGTA
miR-20b-5p CAAAGTGCTCATAGTGCAGGTA
miR-21-5p GGCTAGCTTATCAGACTGATGTT
miR-27a-3p GGCTTCACAGTGGCTAAGTT
miR-29a-3p GTAGCACCATCTGAAATCGGTT
miR-30a-5p GTGTAAACATCCTCGACTGGAA
miR-30c-5p GCTGTAAACATCCTACACTCTCA
miR-30e-5p GGTGTAAACATCCTTGACTGGAA
miR-92a-3p GTATTGCACTTGTCCCGGC
miR-100-5p GAACCCGTAGATCCGAACTT
miR-106b-5p GCTAAAGTGCTGACAGTGCA
miR-125b-5p GCTCCCTGAGACCCTAACTT
miR-128-1-3p GTCACAGTGAACCGGTCTCTT
miR-200b-3p CGGTAATACTGCCTGGTAATGAT
miR-200c-3p CGTAATACTGCCGGGTAATGAT
miR-221-3p GCTACATTGTCTGCTGGGTT
miR-222-3p CGAGCTACATCTGGCTACT

[i] miR, microRNA.

Statistical analysis

The expression levels of all BC, EC and OC-associated miRNAs were determined as mean ΔCq values of the miRNA normalized against the geometric mean of the four housekeeping genes RNU48, miR-16, miR-26b and miR-103. The expression levels of all miRNA types were separately analyzed using a linear model with cell line as the independent variable. The regression coefficients with 95% confidence intervals were tabulated. This led to color coded heatmaps in which red colors indicate strong deviations in the positive direction and blue colors indicate strong deviations in the negative direction from the expression level in the cell line that served as a reference (AN3CA, BT-474 and BT-20). Dark colors correspond to a P<0.00005, and light colors correspond to a P<0.00025. All other comparisons are presented in gray.

Results

miRNA expression profiles of the cell lines

In the present study, the expression levels of 25 BC, EC and OC-associated miRNAs (let-7a, let-7b, let-7d, miR-7, −9, −15b, −17, −19b, −20a, −20b, −21, −27a, −29a, −30a, −30c, −30e, −92a, −100, −106b, −125b, −128.1, −200b, −200c, −221, −222) were quantified in three BC, EC and OC cell lines. The characteristics of each cell line are presented in Table III.

Table III.

Cell line characteristics.

Table III.

Cell line characteristics.

Cell lineBC subtype classificationReceptor statusPrimary tumorOriginOther characteristics(Refs.)
BT474Luminal BER+, PR+/, HER2+Invasive ductal carcinomaBreastKi-67 high, normally endocrine responsive, variable to chemo therapy response, trastuzumab responsive(9094)
SK-BR-3HER2ER, PR, HER2+Invasive ductal carcinomaMetastasis (pleural effusion)Ki-67 high, trastuzumab respon sive, chemotherapy responsive(90,9295)
BT-20 Triple-negativeER, PR, HER2Basal A-like invasive ductal carcinomaBreastDocetaxel responsive, tamoxifen responsive(9497)
Ishikawa LHRH+ER+, PR+, AR+Endometrial adeno-carcinomaEndometrium Tamoxifen-responsive(9698)
EFE-184 ER+, PRCarcinoma (relapse)Endometrium from ascites fluid Tamoxifen-responsive(56,99)
AN3CA ER, PR+Undifferentiated endometrial adenocarcinomaLymph node metastasisNon-steroid response(98101)
SK-OV-3 LHRH Ovarian adenocarcinomaAscites fluidResistance of TNF, cisplatin-responsive, upregulated expression of FGF16 and PITX2(100104)
OAW-42 AdenocarcinomaAscites fluidUpregulated expression of FGF16 and PITX2, tamoxifen responsive(100,102104)

[i] ER, estrogen receptor; PR, progesterone receptor; HER2, human epidermal growth factor receptor 2; Ki-67, marker of proliferation Ki-67; TNF, tumor necrosis factor; FGF16, fibroblast growth factor 16; PITX2, pituitary homeobox 2.

The statistical analyses demonstrated that comparing the three different cell types (AN3CA, BT-474 and BT-20) revealed a range of moderately to highly differentially expressed miRNAs, which exhibited either marked upregulation or downregulation. By clustering miRNAs with respect to their differential expression characteristics, subgroups of miRNAs featuring potential biomarkers to discriminate between BC, OC and EC cells could be created. The expression data clearly revealed a BC-associated miRNA subpanel with significantly distinct expression levels compared with the gynecological tumor types EC and OC (miRs: let-7b, −21, −27a, −30a, −30c, −30e). Consecutively, miRNA clusters with statistical relevance were defined to allow for discrimination between the three tumor entities in a one-versus-one approach (Fig. 1 and Tables SISIII).

miRNAs discriminating BC from OC cells

Expression analyses could determine a subgroup of ten different miRNAs (miRs: let-7b, −21, 30a, −30c, −30e, −27a, −222, −29a, −128.1, −9) that facilitated an expression level-based discrimination between the BC and OC cell types. The notable types included let-7b, miR-21 and the miR-30 family genes, which were uniformly upregulated in OC cells compared with BC cells. For example, compared with AN3CA cells, miR-let-7b was upregulated by a mean value of 5.08 (95% confidence interval, 4.51, 5.65; P<0.001) in SK-OV-3, 8.37 (7.80, 8.94; P<0.001) in OAW-42 cells, and 2.53 (1.96, 3.10; P<0.001) in EFO-27 OC cells (Table SI). In contrast, regression analyses demonstrated no significant difference of miR-let-7b in all investigated BC cell lines (Fig. 2 and Table SII). The expression levels of miR-30a, miR-30c and miR-30e were also increased in all three investigated OC cell lines. Specifically, compared with AN3CA cells, the miR-30a was significantly increased by a mean value of 0.22 (0.21, 0.23; P<0.001) in SK-OV-3, 0.12 (0.10, 0.13; P<0.001) in OAW-42 cells, and 0.43 (0.42, 0.44; P<0.001) in EFO-27 OC cells. The expression levels of miR-30c and miR-30e were also upregulated by a mean value of 0.21 (0.17, 0.25; P<0.001) and 0.07 (0.06, 0.08; P<0.001) in SK-OV-3 cells, 0.12 (0.08, 0.16; P<0.001) and 0.03 (0.02, 0.04; P<0.001) in OAW-42 cells, and 0.50 (0.46, 0.55; P<0.001) and 0.16 (0.15, 0.17; P<0.001) in EFO-27 OC cells (Table SI). No significant differences were identified among all BC cell lines (Table SII). miR-27 and miR-29a exhibited a moderate downregulation in BC cells, with few inconsistent results depending on the cell line comparison (AN3CA or BT-474). By contrast, miR-9 and miR-128.1 exhibited a general moderate downregulation in OC cells compared with BC cells (Fig. 2 and Tables SI and SII).

miRNAs discriminating BC from EC cells

Among the 25 miRNAs evaluated in the present study, six exhibited distinguishing characteristics in regard to BC compared with EC cell expression profiles (miRs: −30a, −30e, −29a, −15b, −200b, −222). While miR-29a, −30a, −30c and −200b were found to be upregulated in EC cells, miR-15b and miR-222 demonstrated downregulated expression levels in comparison to BC cells. For example, miR-200b was upregulated by a mean value of 5.43 (5.27, 5.58; P<0.001) in Ishikawa cells and by 0.97 (0.81, 1.12; P<0.001) in EFE-184 EM cells. Notably, AN3CA cells did not fully comply to the EC-specific expression level trends, which may be explained by cell-specific molecular characteristics (Fig. 3 and Tables SII and SIII).

miRNAs discriminating OC from EC cells

A total of four miRNAs (miR-92a, −106b, −200b, −222) with altered expression levels that may serve a role in the determination of endometrial compared with ovarian malignancies were identified based on this in vitro approach. Upregulated expression levels of miR-92a, −106b and −200b in EC cell types, as well as an upregulation of miR-222 in OC cells may help to mutually distinguish between these tumor types. Compared with AN3CA cells, miR-222 expression was increased by a mean value of 0.66 (0.52, 0.80; P<0.001) in SK-OV-3 cells and by 0.85 (0.71, 0.99; P<0.001) in OAW-42 OC cells. By contrast, a downregulation was identified in two EC cell lines by a mean value of 0.48 (−0.62, −0.34; P<0.001) in Ishikawa cells and by 0.18 (−0.32, −0.04; P=0.018) in EFE-184 EM cells. However, individual cell line-specific differences need to be taken into account in the assessment of tumor type determination of a potential miRNA subpanel with diagnostic power in this regard (Fig. 4 and Tables SI and SIII).

Discussion

The search for clinically applicable biomarkers necessitates a stringent multi-step selection process to singularize, evaluate and validate the usability of a potential biomolecule, or even grouped biomolecule expression profiles or signatures for clear diagnostic purposes. The present study focused on one possible initial step in the determination of potential novel biomarkers that help to detect and distinguish healthy women from patients with malignant disease of the breast, endometrium or ovaries. Based on an in vitro model approach, the proof of principle was accomplished to corroborate the initial hypothesis of discriminating diagnostic features of miRNA signatures in the diagnosis of breast and gynecological malignancies. In general, the detected intracellular miRNA expression levels can be transferred to the extracellular setting of secreted miRNAs, as shown in a previous study (20). Therefore, the experimental design of the present study was targeted on the identification of miRNA signatures, based on tumor-specific expression differences, that enable the mutual discrimination of the three common female cancer types BC, OC and EC. Since miRNAs are robust and easily accessible biomolecules that can be quantified in a wide range of biomaterials, including tissue and liquid biopsies, they meet important requirements for modern and applicable diagnostic biomarkers (44). Thousands of different human miRNAs have been described, of which a clinically relevant subset of 25 different miRNAs with potential impacts in BC, EC and/or OC was pre-selected for the present analytical in vitro approach. Although certain differences in cell line-based and in vivo settings need to be kept in consideration, the current study is intended to provide initial findings that guide further investigations in a promising direction.

Global expression profile analyses in the present study resulted in the identification of cancer type-specific miRNA subgroups. These clusters of distinct miRNAs were characterized by differences in expression levels that can significantly discriminate between the tumor types BC, OC and EC. However, no significant subtype-specific miRNA expression signature differences could be detected among the respective cancer types analyzed.

A parallel comparison of entity-specific clustering habits highlighted a BC-specific miRNA subpanel of six miRNAs that exhibited significantly different expression levels compared with those observed in EC and OC in vitro models. In particularly, members of the miR-30 family were identified in this respect.

Comparisons of miRNA expression signatures in either BC/OC, BC/EC or EC/OC clearly revealed the most miRNA expression profile differences in the comparison of BC vs. OC, with ten of the 25 miRNAs exhibiting significantly different expression levels in these tumor types. Members of the miR-30 family were identified to be significantly differentially expressed, in addition to few more types, including miR-9, which has previously been described as a prognostic marker in OC (45), as well as miR-222 and miR-29a, which are known triggers in breast cancer therapy resistance mechanisms (46). In previous studies, the let-7 family has been reported to exhibit decreased expression levels in OC tissues as well as in OC cell lines, and has been identified to serve a role in OC progression (47,48). In contrast to the literature, in the present study, let-7b was found to be upregulated in OC cells compared with BC cells.

A direct comparison of EC and BC miRNA expression signatures revealed six miRNAs with significantly different expression levels. Again, members of the miR-30 family were prominent, but also miR-15b and mi-200b were identified in this comparison. miR-15b has been described as an aberrantly regulated tumor suppressor (49), whereas miR-200b has a role in epithelial-mesenchymal transition processes (50).

The miRNAs miR-92a, miR-106b, miR-200b and miR-222 compose the smaller subgroup of four miRNAs that exhibited significant expression differences in EC compared with OC in vitro models. Consistent with a previous study by Záveský et al (51), the present data confirmed the differential expression of the miRNAs miR-92a, miR-106b and miR-200b in EC compared with OC. Upregulated expression levels of miR-222 in OC cells were found to associated with epithelial OC in a previous investigation (52).

In conclusion, the diagnostic power and validity of entity-specific miRNA clusters is partially limited due to individual cell type characteristics, such as receptor status or tumor origin (primary tumor or metastasis). For instance, in the present study the estrogen receptor (ER) EC cell line exhibited a different miRNA expression compared with two ER+ EC cell lines. In addition, EFO-27 deviated from the other OC in vitro models to a certain extent, thus an intra-entity variation in miRNA expression signatures has to be taken into account. Furthermore, the present analyses revealed a notable difference in the molecular relationship of EC and OC compared with BC. Therefore, the number of miRNAs with distinguishing expression levels was markedly increased in EC and OC vs. BC than in the comparison between EC vs. OC.

To pursue the identification of a clinically valuable highly entity-specific signature panel with diagnostic power for implementation in routine screening, the obtained data of the present discovery phase approach require further verification and validation. Thus, additional and extended in vitro analyses, followed by translational studies using patients' tissues and liquid biopsy materials should be performed in further analyses to provide substantial evidence for miRNA-based biomarker expression signatures that enable tumor detection, characterization and potential therapy monitoring.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and material

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

The project idea and experimental design was conceived by MH, MJ, DW, SM, BK and TE. In vitro experiments were performed by MJ, CN and DW. Data and statistical analyses were performed by GR, supported by DW, TE and MH. MH, IG, JW, TE, GR, MV, KB and JA interpreted the results and wrote the manuscript. GR, MV, BK, KB and SM critically revised the final version of the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

BC

breast cancer

EC

endometrial cancer

miRNA/miR

microRNA

OC

ovarian cancer

References

1 

Heywang-Koebrunner S, Bock K, Heindel W, Hecht G, Regitz-Jedermann L, Hacker A and Kaeaeb-Sanyal V: Mammography Screening-as of 2013. Geburtshilfe Frauenheilkd. 73:1007–1016. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Clarke MA, Long BJ, Del Mar Morillo A, Arbyn M, Bakkum-Gamez JN and Wentzensen N: Association of endometrial cancer risk with postmenopausal bleeding in Women: A systematic review and meta-analysis. JAMA Intern Med. 178:1210–1222. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Steiner E, Eicher O, Sagemüller J, Schmidt M, Pilch H, Tanner B, Hengstler JG, Hofmann M and Knapstein PG: Multivariate independent prognostic factors in endometrial carcinoma: A clinicopathologic study in 181 patients: 10 years experience at the department of obstetrics and gynecology of the Mainz University. Int J Gynecol Cancer. 13:197–203. 2003. View Article : Google Scholar : PubMed/NCBI

4 

Tejerizo-Garcia A, Jiménez-López JS, Muñoz-González JL, Bartolomé-Sotillos S, Marqueta-Marqués L, López-González G and Gómez JF: Overall survival and disease-free survival in endometrial cancer: Prognostic factors in 276 patients. Onco Targets Ther. 9:1305–1313. 2013.PubMed/NCBI

5 

Das PM and Bast RC Jr: Early detection of ovarian cancer. Biomark Med. 2:291–303. 2008. View Article : Google Scholar : PubMed/NCBI

6 

Howlader N, Noone AM, Krapcho M, Miller D, Brest A, Yu M, Ruhl J, Tatalovich Z, Mariotto A, Lewis DR, Chen HS, Feuer EJ and Cronin KA: SEER Cancer Statistics Review. 1975-2017, National Cancer Institute; Bethesda, MD, USA: https://seer.cancer.gov/csr/1975_2017/

7 

Frangogiannis NG: Biomarkers: Hopes and challenges in the path from discovery to clinical practice. Transl Res. 159:197–204. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Makarova JA, Shkurnikov MU, Wicklein D, Lange T, Samatov TR, Turchinovich AA and Tonevitsky AG: Intracellular and extracellular microRNA: An update on localization and biological role. Prog Histochem Cytochem. 51:33–49. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Hayes J, Peruzzi PP and Lawler S: MicroRNAs in cancer: Biomarkers, functions and therapy. Trends in Molecular Medicine. 20:460–469. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Croce CM: Causes and consequences of microRNA dysregulation in cancer. Nat Rev Genet. 10:704–714. 2009. View Article : Google Scholar : PubMed/NCBI

11 

Kanekura K, Nishi H, Isaka K and Kuroda M: MicroRNA and gynecologic cancers. J Obstet Gynaecol Res. 42:612–617. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Kurozumi S, Yamaguchi Y, Kurosumi M, Ohira M, Matsumoto H and Horiguchi J: Recent trends in microRNA research into breast cancer with particular focus on the associations between microRNAs and intrinsic subtypes. J Hum Genet. 62:15–24. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Nakamura K, Sawada K, Yoshimura A, Kinose Y, Nakatsuka E and Kimura T: Clinical relevance of circulating cell-free microRNAs in ovarian cancer. Mol Cancer. 15:482016. View Article : Google Scholar : PubMed/NCBI

14 

Rapisuwon S, Vietsch EE and Wellstein A: Circulating biomarkers to monitor cancer progression and treatment. Comput Struct Biotechnol J. 14:211–222. 2016. View Article : Google Scholar : PubMed/NCBI

15 

Torres A, Torres K, Pesci A, Ceccaroni M, Paszkowski T, Cassandrini P, Zamboni G and Maciejewski R: Diagnostic and prognostic significance of miRNA signatures in tissues and plasma of endometrioid endometrial carcinoma patients. Int J Cancer. 132:1633–1645. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Widodo, Djati MS and Rifa'i M: Role of microRNAs in carcinogenesis that potential for biomarker of endometrial cancer. Ann Med Surg (Lond. 7:9–13. 2016. View Article : Google Scholar : PubMed/NCBI

17 

Yanokura M, Banno K, Iida M, Irie H, Umene K, Masuda K, Kobayashi Y, Tominaga E and Aoki D: MicroRNAS in endometrial cancer: Recent advances and potential clinical applications. Excli J. 14:190–198. 2015.PubMed/NCBI

18 

Hirschfeld M, Rücker G, Weiß D, Berner K, Ritter A, Jäger M and Erbes T: Urinary exosomal MicroRNAs as potential non-invasive biomarkers in breast cancer detection. Mol Diagn Ther. 24:215–232. 2020. View Article : Google Scholar : PubMed/NCBI

19 

Ritter A, Hirschfeld M, Berner K, Jaeger M, Grundner-Culemann F, Schlosser P, Asberger J, Weiss D, Noethling C, Mayer S and Erbes T: Discovery of potential serum and urine-based microRNA as minimally-invasive biomarkers for breast and gynecological cancer. Cancer Biomark. 27:225–242. 2020. View Article : Google Scholar : PubMed/NCBI

20 

Ritter A, Hirschfeld M, Berner K, Rücker G, Jäger M, Weiss D, Medl M, Nöthling C, Gassner S, Asberger J and Erbes T: Circulating noncoding RNA-biomarker potential in neoadjuvant chemotherapy of triple negative breast cancer? Int J Oncol. 56:47–68. 2020.PubMed/NCBI

21 

Lou Y, Yang X, Wang F, Cui Z and Huang Y: MicroRNA-21 promotes the cell proliferation, invasion and migration abilities in ovarian epithelial carcinomas through inhibiting the expression of PTEN protein. Int J Mol Med. 26:819–827. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Qin X, Yan L, Zhao X, Li C and Fu Y: MicroRNA-21 overexpression contributes to cell proliferation by targeting PTEN in endometrioid endometrial cancer. Oncol Lett. 4:1290–1296. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Frankel LB, Christoffersen NR, Jacobsen A, Lindow M, Krogh A and Lund AH: Programmed cell death 4 (PDCD4) is an important functional target of the microRNA miR-21 in breast cancer cells. J Biol Chem. 283:1026–1033. 2008. View Article : Google Scholar : PubMed/NCBI

24 

Yan LX, Huang XF, Shao Q, Huang MY, Deng L, Wu QL, Zeng YX and Shao JY: MicroRNA miR-21 overexpression in human breast cancer is associated with advanced clinical stage, lymph node metastasis and patient poor prognosis. RNA. 14:2348–2360. 2008. View Article : Google Scholar : PubMed/NCBI

25 

Au Yeung CL, Co NN, Tsuruga T, Yeung TL, Kwan SY, Leung CS, Li Y, Lu ES, Kwan K, Wong KK, et al: Exosomal transfer of stroma-derived miR21 confers paclitaxel resistance in ovarian cancer cells through targeting APAF1. Nat Commun. 7:11502016. View Article : Google Scholar

26 

Greene SB, Herschkowitz JI and Rosen JM: Small players with big roles: microRNAs as targets to inhibit breast cancer progression. Curr Drug Targets. 11:1059–1073. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Encarnacion J, Ortiz C, Vergne R, Vargas W, Coppola D and Matta JL: High DRC Levels are associated with Let-7b overexpression in women with breast cancer. Int J Mol Sci. 17:8652016. View Article : Google Scholar

28 

Chung YW, Bae HS, Song JY, Lee JK, Lee NW, Kim T and Lee KW: Detection of microRNA as novel biomarkers of epithelial ovarian cancer from the serum of ovarian cancer patients. Int J Gynecol Cancer. 23:673–679. 2013. View Article : Google Scholar : PubMed/NCBI

29 

Tang Z, Ow GS, Thiery JP, Ivshina AV and Kuznetsov VA: Meta-analysis of transcriptome reveals let-7b as an unfavorable prognostic biomarker and predicts molecular and clinical subclasses in high-grade serous ovarian carcinoma. Int J Cancer. 134:306–318. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Yu F, Yao H, Zhu P, Zhang X, Pan Q, Gong C, Huang Y, Hu X, Su F, Lieberman J and Song E: let-7 regulates self renewal and tumorigenicity of breast cancer cells. Cell. 131:1109–1123. 2007. View Article : Google Scholar : PubMed/NCBI

31 

Cheng CW, Wang HW, Chang CW, Chu HW, Chen CY, Yu JC, Chao JI, Liu HF, Ding SL and Shen CY: MicroRNA-30a inhibits cell migration and invasion by downregulating vimentin expression and is a potential prognostic marker in breast cancer. Breast Cancer Res Treat. 134:1081–1093. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Rodriguez-Gonzalez FG, Sieuwerts AM, Smid M, Look MP, Meijer-van Gelder ME, de Weerd V, Sleijfer S, Martens JW and Foekens JA: MicroRNA-30c expression level is an independent predictor of clinical benefit of endocrine therapy in advanced estrogen receptor positive breast cancer. Breast Cancer Res Treat. 127:43–51. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Bockhorn J, Dalton R, Nwachukwu C, Huang S, Prat A, Yee K, Chang YF, Huo D, Wen Y, Swanson KE, et al: MicroRNA-30c inhibits human breast tumour chemotherapy resistance by regulating TWF1 and IL-11. Nat Commun. 4:13932013. View Article : Google Scholar : PubMed/NCBI

34 

Lee H, Park CS, Deftereos G, Morihara J, Stern JE, Hawes SE, Swisher E, Kiviat NB and Feng Q: MicroRNA expression in ovarian carcinoma and its correlation with clinicopathological features. World J Surg Oncol. 10:1742012. View Article : Google Scholar : PubMed/NCBI

35 

Cabarcas SM, Thomas S, Zhang X, Cherry JM, Sebastian T, Yerramilli S, Lader E, Farrar WL and Hurt EM: The role of upregulated miRNAs and the identification of novel mRNA targets in prostatospheres. Genomics. 99:108–117. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Wang Y, Li L, Qu Z, Li R, Bi T, Jiang J and Zhao H: The expression of miR-30a* and miR-30e* is associated with a dualistic model for grading ovarian papillary serious carcinoma. Int J Oncol. 44:1904–1914. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Shang C, Lu YM and Meng LR: MicroRNA-125b down-regulation mediates endometrial cancer invasion by targeting ERBB2. Med Sci Moni. 18:BR149–BR155. 2012.

38 

Li C, Gao Y, Zhang K, Chen J, Han S, Feng B, Wang R and Chen L: Multiple Roles of MicroRNA-100 in human cancer and its therapeutic potential. Cellular Physiology and Biochemistry. 37:2143–2159. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Iorio MV, Ferracin M, Liu CG, Veronese A, Spizzo R, Sabbioni S, Magri E, Pedriali M, Fabbri M, Campiglio M, et al: MicroRNA gene expression deregulation in human breast cancer. Cancer research. 65:7065–7070. 2005. View Article : Google Scholar : PubMed/NCBI

40 

Mattie MD, Benz CC, Bowers J, Sensinger K, Wong L, Scott GK, Fedele V, Ginzinger D, Getts R and Haqq C: Optimized high-throughput microRNA expression profiling provides novel biomarker assessment of clinical prostate and breast cancer biopsies. Mol Cancer. 5:242006. View Article : Google Scholar : PubMed/NCBI

41 

Guan Y, Yao H, Zheng Z, Qiu G and Sun K: miR-125b targets BCL3 and suppresses ovarian cancer proliferation. Int J Cancer. 128:2274–2283. 2011. View Article : Google Scholar : PubMed/NCBI

42 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

43 

Pfaffl MW, Tichopad A, Prgomet C and Neuvians TP: Determination of stable housekeeping genes, differentially regulated target genes and sample integrity: BestKeeper-Excel-based tool using pair-wise correlations. Biotechnol Lett. 26:509–515. 2004. View Article : Google Scholar : PubMed/NCBI

44 

Wang H, Peng R, Wang J, Qin Z and Xue L: Circulating microRNAs as potential cancer biomarkers: The advantage and disadvantage. Clin Epigenetics. 10:592018. View Article : Google Scholar : PubMed/NCBI

45 

Sun H, Shao Y, Huang J, Sun S, Liu Y, Zhou P and Yang H: Prognostic value of microRNA-9 in cancers: A systematic review and meta-analysis. Oncotarget. 7:67020–67032. 2016. View Article : Google Scholar : PubMed/NCBI

46 

Zhong S, Li W, Chen Z, Xu J and Zhao J: miR-222 and miR-29a contribute to the drug-resistance of breast cancer cells. Gene. 531:8–14. 2013. View Article : Google Scholar : PubMed/NCBI

47 

Dahiya N and Morin PJ: MicroRNAs in ovarian carcinomas. Endocr Relat Cancer. 17:F77–F89. 2010. View Article : Google Scholar : PubMed/NCBI

48 

Dahiya N, Sherman-Baust CA, Wang TL, Davidson B, Shih IeM, Zhang Y, Wood W III, Becker KG and Morin PJ: MicroRNA expression and identification of putative miRNA targets in ovarian cancer. PLoS One. 3:e24362008. View Article : Google Scholar : PubMed/NCBI

49 

Zhao C, Wang G, Zhu Y, Li X, Yan F, Zhang C, Huang X and Zhang Y: Aberrant regulation of miR-15b in human malignant tumors and its effects on the hallmarks of cancer. Tumour Biol. 37:177–183. 2016. View Article : Google Scholar : PubMed/NCBI

50 

Madhavan D, Peng C, Wallwiener M, Zucknick M, Nees J, Schott S, Rudolph A, Riethdorf S, Trumpp A, Pantel K, et al: Circulating miRNAs with prognostic value in metastatic breast cancer and for early detection of metastasis. Carcinogenesis. 37:461–470. 2016. View Article : Google Scholar : PubMed/NCBI

51 

Záveský L, Jandáková E, Turyna R, Langmeierová L, Weinberger V, Záveská Drábková L, Hůlková M, Hořínek A, Dušková D, Feyereisl J, et al: Evaluation of Cell-Free Urine microRNAs expression for the use in diagnosis of ovarian and endometrial cancers. A Pilot Study. Pathol Oncol Res. 21:1027–1035. 2015. View Article : Google Scholar : PubMed/NCBI

52 

Sun C, Li N, Zhou B, Yang Z, Ding D, Weng D, Meng L, Wang S, Zhou J, Ma D and Chen G: miR-222 is upregulated in epithelial ovarian cancer and promotes cell proliferation by downregulating P27kip1. Oncol Lett. 6:507–512. 2013. View Article : Google Scholar : PubMed/NCBI

53 

Lin Z, Li JW, Wang Y, Chen T, Ren N, Yang L, Xu W, He H, Jiang Y, Chen X, et al: Abnormal miRNA-30e expression is associated with breast cancer progression. Clin Lab. 62:121–128. 2016. View Article : Google Scholar : PubMed/NCBI

54 

Kim SJ, Shin JY, Lee KD, Bae YK, Sung KW, Nam SJ and Chun KH: MicroRNA let-7a suppresses breast cancer cell migration and invasion through downregulation of C-C chemokine receptor type 7. Breast Cancer Res. 14:1–12. 2012. View Article : Google Scholar

55 

Liu K, Zhang C, Li T, Ding Y, Tu T, Zhou F, Qi W, Chen H and Sun X: Let-7a inhibits growth and migration of breast cancer cells by targeting HM. Int J Oncol. 46:2526–2534. 2015. View Article : Google Scholar : PubMed/NCBI

56 

Wang L, Zheng W, Zhang S, Chen X and Hornung D: Expression of monocyte chemotactic protein-1 in human endometrial cancer cells and the effect of treatment with tamoxifen or buserelin. J Int Med Res. 34:284–290. 2006. View Article : Google Scholar : PubMed/NCBI

57 

Chun SM, Park HJ, Kim CH and Kim I: The Significance of MicroRNA Let-7b, miR-30c, and miR-200c Expression in Breast Cancers. J Pathol Transl Med. 45:354–360. 2011.

58 

Liu P, Qi M, Ma C, Lao G and Liu Y and Liu Y and Liu Y: Let7a inhibits the growth of endometrial carcinoma cells by targeting Aurora-B. FEBS Lett. 587:2523–2529. 2013. View Article : Google Scholar : PubMed/NCBI

59 

Bayani J, Kuzmanov U, Saraon P, Fung WA, Soosaipillai A, Squire JA and Diamandis EP: Copy number and expression alterations of miRNAs in the ovarian cancer cell line OVCAR-3: Impact on kallikrein 6 protein expression. Clin Chem. 59:296–305. 2013. View Article : Google Scholar : PubMed/NCBI

60 

Dong P, Ihira K, Xiong Y, Watari H, Hanley SJ, Yamada T, Hosaka M, Kudo M, Yue J and Sakuragi N: Reactivation of epigenetically silenced miR-124 reverses the epithelial-to-mesenchymal transition and inhibits invasion in endometrial cancer cells via the direct repression of IQGAP1 expression. Oncotarget. 7:20260–20270. 2016. View Article : Google Scholar : PubMed/NCBI

61 

Nam EJ, Yoon H, Kim SW, Kim H, Kim YT, Kim JH, Kim JW and Kim S: MicroRNA expression profiles in serous ovarian carcinoma. Clin Cancer Res. 14:2690–2695. 2008. View Article : Google Scholar : PubMed/NCBI

62 

Boren T, Xiong Y, Hakam A, Wenham R, Apte S, Wei Z, Kamath S, Chen DT, Dressman H and Lancaster JM: MicroRNAs and their target messenger RNAs associated with endometrial carcinogenesis. Gynecol Oncol. 110:206–215. 2008. View Article : Google Scholar : PubMed/NCBI

63 

Kowalewska M, Bakula-Zalewska E, Chechlinska M, Goryca K, Nasierowska-Guttmejer A, Danska-Bidzinska A and Bidzinski M: microRNAs in uterine sarcomas and mixed epithelial-mesenchymal uterine tumors: A preliminary report. Tumour Biol. 34:2153–2160. 2013. View Article : Google Scholar : PubMed/NCBI

64 

Markou A, Zavridou M, Sourvinou I, Yousef G, Kounelis S, Malamos N, Georgoulias V and Lianidou E: Direct comparison of metastasis-related miRNAs expression levels in circulating tumor cells, corresponding plasma, and primary tumors of breast cancer patients. Clin Chem. 62:1002–1011. 2016. View Article : Google Scholar : PubMed/NCBI

65 

Bertoli G, Cava C and Castiglioni I: MicroRNAs: New biomarkers for diagnosis, prognosis, therapy prediction and therapeutic tools for breast cancer. Theranostics. 5:1122–1143. 2015. View Article : Google Scholar : PubMed/NCBI

66 

Echevarria-Vargas IM, Valiyeva F and Vivas-Mejia PE: Upregulation of miR-21 in cisplatin resistant ovarian cancer via JNK-1/c-Jun pathway. PLoS One. 9:e970942014. View Article : Google Scholar : PubMed/NCBI

67 

Myatt SS, Wang J, Monteiro LJ, Christian M, Ho KK, Fusi L, Dina RE, Brosens JJ, Ghaem-Maghami S and Lam EW: Definition of microRNAs that repress expression of the tumor suppressor gene FOXO1 in endometrial cancer. 70:367–377. 2010.PubMed/NCBI

68 

Ouzounova M, Vuong T, Ancey PB, Ferrand M, Durand G, Le-Calvez Kelm F, Croce C, Matar C, Herceg Z and Hernandez-Vargas H: MicroRNA miR-30 family regulates non-attachment growth of breast cancer cells. BMC Genomics. 14:1392013. View Article : Google Scholar : PubMed/NCBI

69 

Chang CW, Yu JC, Hsieh YH, Yao CC, Chao JI, Chen PM, Hsieh HY, Hsiung CN, Chu HW, Shen CY and Cheng CW: MicroRNA-30a increases tight junction protein expression to suppress the epithelial-mesenchymal transition and metastasis by targeting Slug in breast cancer. Oncotarget. 7:16462–16478. 2016. View Article : Google Scholar : PubMed/NCBI

70 

Berber U, Yilmaz I, Narli G, Haholu A, Kucukodaci Z and Demirel D: miR-205 and miR-200c: Predictive Micro RNAs for lymph node metastasis in triple negative breast cancer. J Breast Cancer. 17:143–148. 2014. View Article : Google Scholar : PubMed/NCBI

71 

Tsukamoto O, Miura K, Mishima H, Abe S, Kaneuchi M, Higashijima A, Miura S, Kinoshita A, Yoshiura K and Masuzaki H: Identification of endometrioid endometrial carcinoma-associated microRNAs in tissue and plasma. Gynecol Oncol. 132:715–721. 2014. View Article : Google Scholar : PubMed/NCBI

72 

Nagaraja AK, Creighton CJ, Yu Z, Zhu H, Gunaratne PH, Reid JG, Olokpa E, Itamochi H, Ueno NT, Hawkins SM, et al: A link between mir-100 and FRAP1/mTOR in clear cell ovarian cancer. Mol Endocrinol. 24:447–463. 2010. View Article : Google Scholar : PubMed/NCBI

73 

Calura E, Fruscio R, Paracchini L, Bignotti E, Ravaggi A, Martini P, Sales G, Beltrame L, Clivio L, Ceppi L, et al: MiRNA landscape in stage I epithelial ovarian cancer defines the histotype specificities. Clin Cancer Res. 19:4114–4123. 2013. View Article : Google Scholar : PubMed/NCBI

74 

Zhou J, Gong G, Tan H, Dai F, Zhu X, Chen Y, Wang J, Liu Y, Chen P, Wu X and Wen J: Urinary microRNA-30a-5p is a potential biomarker for ovarian serous adenocarcinoma. Oncol Rep. 33:2915–2923. 2015. View Article : Google Scholar : PubMed/NCBI

75 

Ayaz L, Çayan F, Balci Ş, Görür A, Akbayir S, Yıldırım Yaroğlu H, Doğruer Unal N and Tamer L: Circulating microRNA expression profiles in ovarian cancer. J Obstet Gynaecol. 34:620–624. 2014. View Article : Google Scholar : PubMed/NCBI

76 

Hausler SF, Keller A, Chandran PA, Ziegler K, Zipp K, Heuer S, Krockenberger M, Engel JB, Hönig A, Scheffler M, et al: Whole blood-derived miRNA profiles as potential new tools for ovarian cancer screening. Br J Cancer. 103:693–700. 2010. View Article : Google Scholar : PubMed/NCBI

77 

Shukla K, Sharma AK, Ward A, Will R, Hielscher T, Balwierz A, Breunig C, Münstermann E, König R, Keklikoglou I and Wiemann S: MicroRNA-30c-2-3p negatively regulates NF-κB signaling and cell cycle progression through downregulation of TRADD and CCNE1 in breast cancer. Mol Oncol. 9:1106–1119. 2015. View Article : Google Scholar : PubMed/NCBI

78 

Tanic M, Yanowsky K, Rodriguez-Antona C, Andrés R, Márquez-Rodas I, Osorio A, Benitez J and Martinez-Delgado B: Deregulated miRNAs in hereditary breast cancer revealed a role for miR-30c in regulating KRAS oncogene. PLoS One. 7:e388472012. View Article : Google Scholar : PubMed/NCBI

79 

Sorrentino A, Liu CG, Addario A, Peschle C, Scambia G and Ferlini C: Role of microRNAs in drug-resistant ovarian cancer cells. Gynecol Oncol. 111:478–486. 2008. View Article : Google Scholar : PubMed/NCBI

80 

Gong Y, He T, Yang L, Yang G, Chen Y and Zhang X: The role of miR-100 in regulating apoptosis of breast cancer cells. Sci Rep. 5:116502015. View Article : Google Scholar : PubMed/NCBI

81 

Zaman MS, Maher DM, Khan S, Jaggi M and Chauhan SC: Current status and implications of microRNAs in ovarian cancer diagnosis and therapy. J Ovarian Res. 5:442012. View Article : Google Scholar : PubMed/NCBI

82 

Okuda H, Xing F, Pandey PR, Sharma S, Watabe M, Pai SK, Mo YY, Iiizumi-Gairani M, Hirota S, Liu Y, et al: miR-7 suppresses brain metastasis of breast cancer stem-like cells by modulating KLF4. Cancer Res. 73:1434–1444. 2013. View Article : Google Scholar : PubMed/NCBI

83 

Banno K, Yanokura M, Iida M, Adachi M, Nakamura K, Nogami Y, Umene K, Masuda K, Kisu I, Nomura H, et al: Application of microRNA in diagnosis and treatment of ovarian cancer. Biomed Res Int. 2014:2328172014. View Article : Google Scholar : PubMed/NCBI

84 

Ramon LA, Braza-Boïls A, Gilabert J, Chirivella M, España F, Estellés A and Gilabert-Estellés J: microRNAs related to angiogenesis are dysregulated in endometrioid endometrial cancer. Hum Reprod. 27:3036–3045. 2012. View Article : Google Scholar : PubMed/NCBI

85 

Kinose Y, Sawada K, Nakamura K and Kimura T: The Role of MicroRNAs in Ovarian Cancer. Biomed Res Int. 2014:112014. View Article : Google Scholar

86 

Lu J, Zhang X, Zhang R and Ge Q: MicroRNA heterogeneity in endometrial cancer cell lines revealed by deep sequencing. Oncol Lett. 10:3457–3465. 2015. View Article : Google Scholar : PubMed/NCBI

87 

Wu Q, Guo L, Jiang F, Li L, Li Z and Chen F: Analysis of the miRNA-mRNA-lncRNA networks in ER+ and ER- breast cancer cell lines. J Cell Mol Med. 19:2874–2887. 2015. View Article : Google Scholar : PubMed/NCBI

88 

Chong GO, Jeon HS, Han HS, Son JW, Lee YH, Hong DG, Lee YS and Cho YL: Differential MicroRNA Expression Profiles in Primary and Recurrent Epithelial Ovarian Cancer. Anticancer Res. 35:2611–2617. 2015.PubMed/NCBI

89 

Singh SR and Rameshwar P: MicroRNA in development and in the progression of cancer. Springer-Verlag; New York: 2014, View Article : Google Scholar

90 

Brockhoff G, Heckel B, Schmidt-Bruecken E, Plander M, Hofstaedter F, Vollmann A and Diermeier S: Differential impact of Cetuximab, Pertuzumab and Trastuzumab on BT474 and SK-BR-3 breast cancer cell proliferation. Cell Prolif. 40:488–507. 2007. View Article : Google Scholar : PubMed/NCBI

91 

Lasfargues EY, Coutinho WG and Redfield ES: Isolation of two human tumor epithelial cell lines from solid breast carcinomas. J Natl Cancer Inst. 61:967–978. 1978.PubMed/NCBI

92 

Subik K, Lee JF, Baxter L, Strzepek T, Costello D, Crowley P, Xing L, Hung MC, Bonfiglio T, Hicks DG and Tang P: The Expression Patterns of ER, PR, HER2, CK5/6, EGFR, Ki-67 and AR by immunohistochemical analysis in breast cancer cell lines. Breast Cancer. 4:35–41. 2010.PubMed/NCBI

93 

Lee S, Yang W, Lan KH, Sellappan S, Klos K, Hortobagyi G, Hung MC and Yu D: Enhanced sensitization to taxol-induced apoptosis by herceptin pretreatment in ErbB2-overexpressing breast cancer cells. Cancer Res. 62:5703–5710. 2002.PubMed/NCBI

94 

Kim DJ, Lee WY, Park NW, Kim GS, Lee KM, Kim J, Choi MK, Lee GH, Han W and Lee SK: Drug response of captured BT20 cells and evaluation of circulating tumor cells on a silicon nanowire platform. Biosens Bioelectron. 67:370–378. 2015. View Article : Google Scholar : PubMed/NCBI

95 

Kloten V, Schlensog M, Eschenbruch J, Gasthaus J, Tiedemann J, Mijnes J, Heide T, Braunschweig T, Knüchel R and Dahl E: Abundant NDRG2 expression is associated with aggressiveness and unfavorable patients' outcome in basal-like breast cancer. PLoS One. 11:e01590732016. View Article : Google Scholar : PubMed/NCBI

96 

Croxtall JD, Elder MG and White JO: Hormonal control of proliferation in the Ishikawa endometrial adenocarcinoma cell line. J Steroid Biochem. 35:665–669. 1990. View Article : Google Scholar : PubMed/NCBI

97 

Nishida M, Kasahara K, Kaneko M, Iwasaki H and Hayashi K: Establishment of a new human endometrial adenocarcinoma cell line, Ishikawa cells, containing estrogen and progesterone receptors. Nihon Sanka Fujinka Gakkai zasshi. 37:1103–1111. 1985.(In Japanese). PubMed/NCBI

98 

Dawe CJ, Banfield WG, Morgan WD, Slatick MS and Curth HO: Growth in continuous culture, and in hamsters, of cells from a neoplasm associated with acanthosis nigricans. J Natl Cancer Inst. 33:441–456. 1964.PubMed/NCBI

99 

Korch C, Spillman MA, Jackson TA, Jacobsen BM, Murphy SK, Lessey BA, Jordan VC and Bradford AP: DNA profiling analysis of endometrial and ovarian cell lines reveals misidentification, redundancy and contamination. Gynecol Oncol. 127:241–248. 2012. View Article : Google Scholar : PubMed/NCBI

100 

Basu M, Mukhopadhyay S, Chatterjee U and Roy SS: FGF16 promotes invasive behavior of SKOV-3 ovarian cancer cells through activation of mitogen-activated protein kinase (MAPK) signaling pathway. J Biol Chem. 289:1415–1428. 2014. View Article : Google Scholar : PubMed/NCBI

101 

Irmer G, Bürger C, Müller R, Ortmann O, Peter U, Kakar SS, Neill JD, Schulz KD and Emons G: Expression of the messenger RNAs for luteinizing hormone-releasing hormone (LHRH) and its receptor in human ovarian epithelial carcinoma. Cancer Res. 55:817–822. 1995.PubMed/NCBI

102 

Yoon J, Kim ES, Lee SJ, Park CW, Cha HJ, Hong BH and Choi KY: Apoptosis-related mRNA expression profiles of ovarian cancer cell lines following cisplatin treatment. J Gynecol Oncol. 21:255–261. 2010. View Article : Google Scholar : PubMed/NCBI

103 

Langdon SP and Lawrie SS: Establishment of ovarian cancer cell lines. Methods Mol Med. 39:155–159. 2001.PubMed/NCBI

104 

Shukla J, Sharma U, Kar R, Varma IK, Juyal S, Jagannathan NR and Bandopadhyaya GP: Tamoxifen-2-hydroxylpropyl-beta-cyclodextrin-aggregated nanoassembly for nonbreast estrogen-receptor-positive cancer therapy. Nanomedicine (Lond). 4:895–902. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2020
Volume 22 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Hirschfeld M, Ge I, Rücker G, Waldschmidt J, Mayer S, Jäger M, Voigt M, Kammerer B, Nöthling C, Berner K, Berner K, et al: Mutually distinguishing microRNA signatures of breast, ovarian and endometrial cancers in vitro. Mol Med Rep 22: 4048-4060, 2020
APA
Hirschfeld, M., Ge, I., Rücker, G., Waldschmidt, J., Mayer, S., Jäger, M. ... Erbes, T. (2020). Mutually distinguishing microRNA signatures of breast, ovarian and endometrial cancers in vitro. Molecular Medicine Reports, 22, 4048-4060. https://doi.org/10.3892/mmr.2020.11466
MLA
Hirschfeld, M., Ge, I., Rücker, G., Waldschmidt, J., Mayer, S., Jäger, M., Voigt, M., Kammerer, B., Nöthling, C., Berner, K., Weiss, D., Asberger, J., Erbes, T."Mutually distinguishing microRNA signatures of breast, ovarian and endometrial cancers in vitro". Molecular Medicine Reports 22.5 (2020): 4048-4060.
Chicago
Hirschfeld, M., Ge, I., Rücker, G., Waldschmidt, J., Mayer, S., Jäger, M., Voigt, M., Kammerer, B., Nöthling, C., Berner, K., Weiss, D., Asberger, J., Erbes, T."Mutually distinguishing microRNA signatures of breast, ovarian and endometrial cancers in vitro". Molecular Medicine Reports 22, no. 5 (2020): 4048-4060. https://doi.org/10.3892/mmr.2020.11466