Identification of long non‑coding RNA expression patterns useful for molecular‑based classification of type I endometrial cancers

  • Authors:
    • Maria Ravo
    • Angela Cordella
    • Pasquale Saggese
    • Antonio Rinaldi
    • Maria Antonietta Castaldi
    • Giovanni Nassa
    • Giorgio Giurato
    • Fulvio Zullo
    • Alessandro Weisz
    • Roberta Tarallo
    • Francesca Rizzo
    • Maurizio Guida
  • View Affiliations

  • Published online on: November 21, 2018     https://doi.org/10.3892/or.2018.6880
  • Pages: 1209-1217
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Endometrial cancer is the most frequently diagnosed gynecologic malignant disease. Although several genetic alterations have been associated with the increased risk of endometrial cancer, to date, the diagnosis and prognosis still rely on morphological features of the tumor, such as histological type, grading and invasiveness. As molecular‑based classification is desirable for optimal treatment and prognosis of these cancers, we explored the potential of lncRNAs as molecular biomarkers. To this end, we first identified by RNA sequencing (RNA‑Seq) a set of lncRNAs differentially expressed in cancer vs. normal endometrial tissues, a result confirmed also by analysis of normal and cancerous endometrium RNA‑Seq data from TCGA (The Cancer Genome Atlas). A significant association of a subset of these differentially expressed lncRNAs with tumor grade was then determined in 405 TCGA endometrial cancer profiles. Integrating endometrial cancer‑specific expression profiles of long and small non‑coding RNAs, a functional association network was then identified. These results describe for the first time a functional ῾core᾽ network, comprising small and long RNAs, whose deregulation is associated with endometrial neoplastic transformation, representing a set of cancer biomarkers that can be monitored and targeted for diagnosis, follow‑up and therapy of these tumors.

Introduction

Endometrial cancer (EC) is the most common malignancy of the female genital tract, with an estimated incidence of 63,230 new cases in 2018 in the United States (1). With the increasing prevalence of major EC risk factors, such as obesity, diabetes and hypertension, a rising trend has also been recognized in younger women (2). EC is generally classified into two subtypes, defined as type I or endometrioid endometrial cancer (EEC) and type II or non-endometrioid endometrial cancer (NEEC) (3,4). This dualistic classification underlies different patterns of molecular alterations, pathogenesis and clinical outcome (4).

Type I EC, which is the most frequent uterine malignancy (80% of all cases), is an estrogen-dependent lesion, often seen in conjunction with endometrial hyperplasia, usually developed in peri- and early postmenopausal women (4). Type II EC affects older patients, is not preceded by hyperplasia and comprises more aggressive histologic subtypes, such as papillary serous, clear cell carcinomas and carcinosarcomas (3,5). In general, type I EC has a good prognosis, with a 10-year overall survival rate exceeding 80% (6,7). Surprisingly, despite optimal risk-adapted treatment, a small but substantial number of patients exhibits recurrence and poor survival. In such cases available risk factors or biological markers are not able to reliably predict the poor clinical course. Indeed, it is currently established that a classification only based on morphologic features is inconsistent and that molecular-based classification is desirable for optimal treatment and prognosis of such cancers. Nevertheless, although several genetic alterations have been associated with increased risk of EC (810) and mutations in genes such as ATR have been established to be associated with poor clinical outcomes in EEC (11), new molecular markers need to be identified for early diagnosis and treatment purposes.

In 2013, researchers at The Cancer Genome Atlas (TCGA) performed an integrated genomic, transcriptomic and proteomic characterization of 373 endometrial carcinomas by applying array- and sequencing-based technologies, thus drawing a new classification of endometrial cancers according to four main molecular categories according to their genomic features (12). This may guide post-surgical therapy in patients affected with aggressive tumors. Yet, since taking these results into clinical practice is currently cost-prohibitive, molecular surrogates corresponding to each of the subgroups (13), able to successfully classify all patients in the TCGA cohort and to minimize false negatives in the CN-high poor prognosticator group, were identified.

The Encyclopedia of DNA Elements (ENCODE) project estimates that almost 62–75% of the DNA is transcribed into RNA, but only 2% of the transcriptome is finally translated into proteins (14). However, this non-coding part of the genome plays many key roles in several biological processes and diseases such as in cancers (15). Thus, the landscape of non-coding RNAs (ncRNAs) is increasing day by day and has emerged as a major source of biomarkers (16,17), being considered as priority transcripts to be monitored for functional significance in both pathogenesis and progression of ECs. Based on their size, ncRNAs are divided in two main categories: Small ncRNAs [sncRNAs (<200 nt)] and long ncRNAs [lncRNAs (>200 nt)]. Moreover, they are connected to each other, with some lncRNAs acting on post-transcriptional regulation through the modulation of certain sncRNA subgroups (18).

Among sncRNAs, four major functional groups have been identified in mammals: microRNAs (miRNAs), PIWI-interacting-RNAs (piRNAs), small nucleolar-RNAs (snoRNAs) and endogenous small interfering-RNAs (endo-siRNAs) (19). sncRNAs can exert large-scale and diverse effects on cellular processes by regulating gene expression, protein translation and genomic organization. In particular, lncRNAs may contain also miRNA-binding sites and can compete with miRNAs for interaction with their target mRNAs and thereby block their effects on these mRNAs.

The classification of lncRNAs is more complex than that of sncRNAs, as they can be divided on the basis of their size, association with annotated protein-coding genes, or with other DNA elements of known function (18,20). Thus, lncRNAs represent a heterogeneous group of RNAs, which have been identified as important molecules in several biological processes, performing tumor suppression and oncogenic functions in various types of cancer (18).

Using small RNA sequencing and microarrays, we previously determined significant differences in sncRNA expression patterns between normal, hyperplastic and neoplastic endometrium of patients affected with EEC (21). This led to the definition of a sncRNA signature (129 miRNAs, 2 of which were not previously described, 10 piRNAs and 3 snoRNAs) recapitulating neoplastic transformation.

In the present study, we aimed to analyze the role of lncRNAs in EEC and to examine a possible connection with the sncRNAs previously observed. To this aim, we first performed lncRNA profiling in a set of EEC samples and paired normal tissues, to identify lncRNA molecules more likely to be associated with the cancerous state. These were also confirmed in a set of 20 paired cancer-normal profiles deposited in the TCGA database. The 80 more highly differentially expressed were found to be significantly associated with different tumor grade among 405 EEC RNA profiles in TCGA. Finally, we identified a small/long RNA functional core whose deregulation was predicted to be strongly associated with endometrial neoplastic transformation. Indeed, computational analysis identified, among the most affected functional pathways, focal adhesion, MAPK and Wnt signaling, confirming previously published data (21).

Materials and methods

Patients and tissue collection

Six patients, who underwent type A radical hysterectomy with or without pelvic lymphadenectomy for EC, were enrolled in the present study at the University of Salerno. Samples were collected from December 2013 to November 2014 in the Division of Gynecology and Obstetrics, ‘SS. Giovanni di Dio e Ruggi d'Aragona’ University Hospital of Salerno (Italy). Average age of patients was 64 (range, 51–80 years). The study protocol received institutional review board (IRB) approval before the beginning of the study, in accordance with the code of ethics of the Declaration of Helsinki, and informed consent was obtained from all patients.

From each patient a sample of neoplastic tissue and corresponding normal endometrium were collected, washed and stored at −80°C until analysis. The histology of each EC was classified according to World Health Organization criteria, whereas surgical staging followed FIGO (International Federation of Gynaecology and Obstetrics) standards.

RNA isolation and quality controls

Tissue specimen were disrupted and homogenized using TissueLyser LT (Qiagen, Hilden, Germany). Total RNA was extracted with miRVana RNA Isolation kit (Thermo Fisher Scientific, Inc., Waltham, MA, USA) according to the manufacturer's protocol. Before use, the RNA concentration in each sample was assayed with a NanoDrop ND-2000c (Thermo Fisher Scientific, Inc.) and its quality was assessed with the Agilent 2100 Bioanalyzer using Agilent RNA 6000 Nano kit (Agilent Technologies, Inc., Santa Clara, CA, USA).

RNA sequencing and data analysis

RNA-Seq libraries were prepared as previously described (22). Briefly, 1 µg of total RNA was used in a library preparation according to the Illumina TruSeq Stranded Total RNA Sample Preparation protocol (Illumina, Inc., San Diego, CA, USA). Each library was sequenced on HiSeq 2500 (Illumina) at a concentration of 8 pM for 200 cycles plus 7 additional cycles for index sequencing in the paired-mode (2×100 base pair).

RNA-Seq data analysis was performed as described in the study by Tarallo et al (23). In details, fastQ underwent to quality control using FastQC tool [http://www.bioinformatics.babraham.ac.uk/projects/fastqc/]. The mapping of paired-end reads was performed on reference genome assembly hg19 using STAR (version 2.5.2b) (24). The quantification of lncRNAs expressed for each sample was performed using HTSeq-Count algorithm (25), while differential expression analysis was performed with DESeq2 (26). Only lncRNAs showing a cut-off of |fold change| ≥1.5 and adjusted P-value (P-adj) ≤0.05 were considered for further analysis.

TCGA raw data, corresponding to either 20 tumor and paired normal or 405 EEC tissues, were analyzed as described above and compared to the in-house investigated samples.

Bioinformatic analyses

Pearson correlation between in-house differentially expressed lncRNAs and paired tumor-normal TCGA data was computed using R software (https://www.r-project.org/).

Non-negative matrix factorization (NMF) was computed considering the normalized expression values in endometrial cancer data taken from TCGA, considering the list of in-house differentially expressed lncRNAs showing |fold change| ≥3 and P-adj ≤0.05 using the R package ‘nmf’ available in CRAN (27).

Kaplan-Meier survival analysis and Cox proportional-hazard regression was generated using MedCalc 18.5 software (https://www.medcalc.org/). lncRNA and mRNA targets of the selected miRNAs were computed using miRWalk 2.0 (28). The miRNA, lncRNA and mRNA associated competing endogenous RNA (ceRNA) network was designed as described by the study of Wang et al (29). Gene Ontology analysis was performed using Co-LncRNA tool (30).

Results

Long non-coding RNA profiling in EEC tissues

lncRNA expression profiling was performed by next-generation sequencing in samples of type I endometrial cancer (EEC), to evaluate their possible deregulation during carcinogenesis.

To this aim, six patients were selected out of a larger cohort following initial characterization according to defined clinicopathological parameters (Table I). For each patient, two endometrial biopsies were obtained from pathological and adjacent normal tissue (normal tissues indicated as: 3N, 4N, 14N, 15N, 18N and 19N; tumor tissue samples indicated as: 3T, 4T, 14T, 15T, 18T and 19T). Normal tissues were collected as far as possible from the area presenting a cancerous lesion, to reduce the possibility of cross-contamination.

Table I.

Clinicopathological features of the tissue samples analyzed from 6 patients affected with endometrioid endometrial cancer.

Table I.

Clinicopathological features of the tissue samples analyzed from 6 patients affected with endometrioid endometrial cancer.

CharacteristicsNPatient nos.
Age (years)
  >5553, 14, 15, 18, 19
  ≤5514
Tissue categories
  Normal63N, 4N, 14N, 15N, 18N, 19N
  Endometrial cancer63T, 4T, 14T, 15T, 18T, 19T
Stage (FIGO)
  I–II53T, 4T, 14T, 15T, 18T
  III–IV119T
Histological grade
  G134T, 14T, 15T
  G213T
  G3218T, 19T
Lymph node metastasis
  Positive0
  Negative53T, 4T, 15T, 18T, 19T,
  Nx114T
Invasion
  T1 and T253T, 4T, 14T, 15T, 18T
  T3 and T4119T

More than 200 million sequences were obtained by RNA-Seq analysis for the 12 samples sequenced, that after filtering out low quality reads and trimming the adaptors led to approximately 20 million reads/sample.

The obtained reads were aligned against the human genome reference (hg19), paying particular care in lncRNA identification. lncRNA molecules included in the human genome are classified in seven categories [long intergenic non-coding RNA (lincRNA), antisense, sense_overlapping, processed_transcript, sense_intronic, bidirectional lncRNA and miRNA] according to GENCODE gene annotation, the largest manually curated catalog of human lncRNAs (31). More than 24,000 RNA molecules, considering both coding and non-coding ones, were identified in total in the investigated samples and the percentage of lncRNAs belonging to the aforementioned categories is reported in Fig. 1A. In particular, most of the lncRNAs expressed within the investigated samples belonged to two main categories: lincRNAs (41%) and antisense transcripts (37%).

Then, since deregulation of lncRNAs has been associated with a broad range of physiological defects in multiple diseases, including cancer, to investigate whether lncRNA expression patterns may change during endometrial carcinogenesis, differential expression analysis was performed (tumor vs. normal) and a pool of lncRNAs specifically deregulated in cancerous tissues was identified.

In particular, 131 lncRNAs (Fig. 1B) differentially expressed between EEC samples and the corresponding normal endometrium were found considering |fold change (FC)| ≥1.5 and P-adj ≤0.05, suggesting their involvement in endometrial carcinogenesis. The full list of statistically significant differentially expressed RNAs is available at ArrayExpress (E-MTAB-7041).

To identify the most relevant lncRNAs in the development of EEC, we performed genome-wide analysis by comparing our data with TCGA molecular RNA-Seq profiles of 20 patients for which data from primary EEC tumors and the corresponding normal endometrial tissues were available. By analyzing the expression of the 131 lncRNAs identified in our patients to be deregulated (|FC| ≥1.5 and P-adj ≤0.05), it emerged that most of them had the same trend in the TCGA dataset, discriminating between cancer and normal tissues (Fig. 1C). In particular, comparing their expression levels, we observed a 0.73 correlation between the two datasets (Fig. 1D). It is also interesting to observe that the lncRNAs having an opposite trend between the two sets of samples were mostly those showing little changes among our samples and being not statistically significant in the TCGA group.

To confirm the consistency of our results, we investigated in particular the behavior of 80 lncRNAs whose expression levels were strongly modulated in tumor samples (|FC| ≥3 and P-adj ≤0.05) among the molecular RNA-Seq profiles of 405 primary EEC tumors deposited in TCGA database.

Thus, we performed integrative analysis in order to identify molecularly distinct tumor EEC subgroups associated with specific clinico-biological features. Unsupervised hierarchical clustering revealed five clusters of lncRNAs that correlated with specific clinical parameters. In particular, the most statistically relevant and discriminating factor resulted in histological grading (P<0.01 by Chi-square test) (Fig. 2A).

Moreover, these five clusters hold statistically different weight on survival (P=0.5890), identifying five subgroups of EECs with slightly different trends in overall survival, as shown in the Kaplan-Meier survival curve of Fig. 2B. Additionally, we analyzed the survival of patients separately in each of the five groups using clinical stage and grade as covariates by applying Cox proportional-hazards regression model. In this way, we found that clusters 1 and 4 were significantly associated with survival in patients at stage 3 (P<0.0228 and P<0.0498, respectively), while survival was strongly associated in patients belonging to cluster 5 classified in stage 4 (P<0.0022). All these patients showed a significantly worse survival.

mRNA-lncRNA co-expression and ceRNA network construction

To better investigate the functional significance of our observations, we correlated the 80 most deregulated lncRNAs with mRNA profiles obtained in the tumor tissues analyzed here. This led to the identification of a set of pathways specifically affected by mRNA-lncRNA co-expression, including MAPK, JAK/Stat and TGF-β signaling (Fig. 2C) that was found also to be affected by sncRNAs in our previous study (21).

Indeed, by integrating these data with previous results from studies of sncRNAs in EEC and considering the possible interplay between miRNAs-mRNAs-lncRNAs, a ceRNA network was obtained, revealing miRNAs specifically targeting either mRNAs or lncRNAs differentially expressed in EEC. To this aim, the list of 129 miRNAs representing a signature of endometrial neoplastic transformation was filtered applying P-value correction (Bonferroni correction), leading to 11 more significant miRNAs. An integrative network was then drawn to show miRNA-mRNA-lncRNA correlations (Fig. 3). In details, the analysis was focused on miRNAs, thus, showing mRNA and lncRNA targets having a negative correlation with respect to miRNA expression. In this context, the most significantly affected functional pathways were found, among others, focal adhesion, ECM receptor interaction, MAPK and Wnt signaling (Fig. 3), suggesting that specific small/long transcripts combination may be directly involved in EEC onset and progression and thus being key markers to be monitored in diagnosis and follow-up of the disease.

Discussion

Molecular classification of ECs represents an objective that clinicians are trying to achieve in order to provide independent prognostic information beyond established risk factors.

In the past few years, pragmatic molecular classifiers have been developed, able to identify four prognostically distinct molecular subgroups that may change the current risk stratification systems (12,13,32). Indeed, the evolution of genomic classification in ECs has the potential to be used routinely in therapeutic protocol selection and in stratifying cases in future clinical trials (13).

Nevertheless, mounting evidence has emphasized the roles and clinical significance of non-coding RNAs and, in particular lncRNAs, among others in endometrial cancer (33,34).

Several studies have shown the potential of lncRNAs as therapeutic targets and investigated their involvement in cancer pathogenesis (35,36). Indeed, limited information is available on EC (3744). Clarification of their role in the onset, progression and follow-up of this tumor is warranted to help molecular classification and targeted therapy of this cancer histotype.

In the present study, we first evaluated the differential expression of lncRNAs between endometrial tumor samples and paired normal tissues of six EEC patients and then we compared and validated our results with different EEC datasets present in TCGA database. In this way, we identified a set of lncRNAs discriminating between normal and cancerous endometrium, common between those described here and publicly available data. Moreover, deep investigation of this lncRNAs dataset among a group of 405 EEC RNA-Seq profiles in TGCA revealed that 80 lncRNAs, differentially expressed with a higher fold, were statistically distributed in five clusters, reflecting tumor grade stratification and associated with different survival trends.

A number of studies have demonstrated that a large number of miRNA-binding sites are present on lncRNAs, suggesting how these transcripts can serve also as competing endogenous RNAs controlling ‘free’ miRNA levels and functions (45,46). Importantly, lncRNAs could compete with mRNAs for miRNAs, and thereby regulate miRNA-mediated transcript repression (45,46). Therefore, we built a ceRNA network, depicting key interactions occurring among differentially expressed mRNAs, lncRNAs and miRNAs in our EEC samples and determining a functional impact within crucial pathways in EC, which may represent potential core regulators to be monitored for early diagnosis and therapeutic purposes. Indeed, understanding molecular interactions has becoming a common tool especially in cancer research, given the limited information provided to date by studies focusing on a single molecule (4749).

In this regard, our results strengthen and integrate the current knowledge concerning EEC molecular markers, thus providing novel details about their functional correlations. In this way, a functional multi-molecule core has been identified, showing miRNA members mostly deregulated in EEC and the corresponding mRNA and lncRNA targets inversely correlated with miRNA expression. From this snapshot it emerges, for example, the coexistence between downregulation of miR-10b that has been also proved to be downregulated in other types of cancer, such as gastric and cervical ones (50,51) and upregulation of its long-RNA targets. Moreover, we confirmed upregulation of almost the entire miR-200 family (miR-200a/b/c and miR-141), an event already well documented in EEC, where these RNAs target mainly genes involved in tissue transformation responsible for epithelial-to-mesenchymal transition (EMT) (52,53). In this context, we observed an inverse correlation, among others, with the well known miR-200 targets ZEB1 and ZEB2, transcriptional repressors of E-cadherin whose expression is restored in EMT (53) and KLF9, whose downregulation in EEC has been previously demonstrated (54,55). Downregulation was also retrieved for TIMP2 representing, together with the above mentioned mRNAs, a key gene involved in the differentiation of EEC and proposed as a potential therapeutic target (5658). More interestingly, an inverse correlation was also found with several downregulated lncRNAs, including ADAMTS9-AS2, MEG3 and HAND2-AS1, already demonstrated to act as tumor suppressors. Indeed, ADAMTS9-AS2 has been associated with the inhibition of cancer progression and migration in lung and glioma cells; its expression has been correlated with poor prognosis through a mechanism involving the interaction with DNMT1 (59,60). In addition, a role of MEG3 in EEC tumorigenesis and progression through the modulation of Notch and PI3K pathways has been proved (40,61). In several cancer types, MEG3 has been shown to sequester various microRNAs from protein and/or target mRNAs resulting in altered protein activity, translation and degradation; in particular involvement of this lncRNA in the regulation of the miR-200 family has been demonstrated, confirming what was revealed by the ceRNA model constructed here (Fig. 3) (62). Finally, HAND2-AS1 has been shown to be involved in inhibition of EEC invasion and metastasis, through the downregulation of neuromedin U (63), and it has been proposed to play a role in the tumorigenesis of muscle-invasive bladder cancer through the interaction with many several miRNAs (including, of note, also miR-183) (64).

In conclusion, in the present study we identified a functional core, constituted by a specific combination of miRNAs-mRNAs-lncRNAs expression, which represents a molecular signature of potential usefulness to monitor EEC progression, for follow-up and prognosis of this disease.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Italian Ministry of Education University and Research (Flagship Project InterOmics), the Italian Association for Cancer Research (AIRC, Grant IG-17426), the University of Salerno (FARB) and the Genomix4Life Srl.

Availability of data and materials

The results shown here are in part based upon data generated by the TCGA Research Network: http://cancergenome.nih.gov/. The datasets generated and analyzed in this study are available in the EBI ArrayExpress database (http://www.ebi.ac.uk/arrayexpress) with Accession Number E-MTAB-7039 (raw data) and E-MTAB-7041.

Authors' contributions

AW, FR, FZ and MG designed and coordinated the study; MAC, AC and MR selected the patients and MAC collected the clinical samples; AC, FR, GN, MR, PS and RT carried out the experimental work; AR and GG performed the data analyses; AC, MR and RT wrote the manuscript; MR, AC, PS, AR, MAC, GN, GG, FZ, AW, RT, FR and MG contributed to data interpretation and discussion and to manuscript revision. All authors read and approved the manuscript and agree to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Ethics approval and consent to participate

The study protocol received approval by the Ethics Committee of the ‘SS. Giovanni di Dio e Ruggi d'Aragona’ University of Salerno Hospital (n.er 91/13.12.2013) before the beginning of the study, in accordance with The Code of Ethics of the Declaration of Helsinki, and informed consent was obtained from all patients included.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Authors' information

Pasquale Saggese is a PhD student of the Research Doctorate in ‘Biomedical Science and Technology’ (XXXI cycle), ‘Roma TRE’ University.

Glossary

Abbreviations

Abbreviations:

EC

endometrial cancer

EEC

endometrioid endometrial cancer

EMT

epithelial-to-mesenchymal transition

FC

fold change

lincRNA

long intergenic non-coding RNA

lncRNA

long non-coding RNA

miRNA

microRNA

ncRNA

non-coding RNA

ceRNA

competing endogenous RNA

NEEC

non-endometrioid endometrial cancer

P-adj

adjusted P-value

piRNA

PIWI-interacting RNA

sncRNA

small non-coding RNA

TCGA

The Cancer Genome Atlas

References

1 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2018. CA Cancer J Clin. 68:7–30. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Siegel R, Ma J, Zou Z and Jemal A: Cancer statistics, 2014. CA Cancer J Clin. 64:9–29. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Koh WJ, Greer BE, Abu-Rustum NR, Apte SM, Campos SM, Chan J, Cho KR, Cohn D, Crispens MA, Dupont N, et al: Uterine neoplasms, version 1.2014. J Natl Compr Canc Netw. 12:248–280. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Zannoni GF, Scambia G and Gallo D: The dualistic model of endometrial cancer: The challenge of classifying grade 3 endometrioid carcinoma. Gynecol Oncol. 127:262–263. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Amant F, Cadron I, Fuso L, Berteloot P, de Jonge E, Jacomen G, Van Robaeys J, Neven P, Moerman P and Vergote I: Endometrial carcinosarcomas have a different prognosis and pattern of spread compared to high-risk epithelial endometrial cancer. Gynecol Oncol. 98:274–280. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Lajer H, Elnegaard S, Christensen RD, Ortoft G, Schledermann DE and Mogensen O: Survival after stage IA endometrial cancer; can follow-up be altered? A prospective nationwide Danish survey. Acta Obstet Gynecol Scand. 91:976–982. 2012. View Article : Google Scholar : PubMed/NCBI

7 

Kitchener HC and Trimble EL: Endometrial Cancer Working Group of the Gynecologic Cancer Intergroup: Endometrial cancer state of the science meeting. Int J Gynecol Cancer. 19:134–140. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Torricelli F, Nicoli D, Bellazzi R, Ciarrocchi A, Farnetti E, Mastrofilippo V, Zamponi R, La Sala GB, Casali B and Mandato VD: Computational development of a molecular-based approach to improve risk stratification of endometrial cancer patients. Oncotarget. 9:25517–25528. 2018. View Article : Google Scholar : PubMed/NCBI

9 

Lee PJ, McNulty S, Duncavage EJ, Heusel JW and Hagemann IS: Clinical targeted next-generation sequencing shows increased mutational load in endometrioid-type endometrial adenocarcinoma with deficient DNA mismatch repair. Int J Gynecol Pathol. 37:581–589. 2017.

10 

DeLair DF, Burke KA, Selenica P, Lim RS, Scott SN, Middha S, Mohanty AS, Cheng DT, Berger MF, Soslow RA and Weigelt B: The genetic landscape of endometrial clear cell carcinomas. J Pathol. 243:230–241. 2017. View Article : Google Scholar : PubMed/NCBI

11 

Zighelboim I, Schmidt AP, Gao F, Thaker PH, Powell MA, Rader JS, Gibb RK, Mutch DG and Goodfellow PJ: ATR mutation in endometrioid endometrial cancer is associated with poor clinical outcomes. J Clin Oncol. 27:3091–3096. 2009. View Article : Google Scholar : PubMed/NCBI

12 

Cancer Genome Atlas Research Network, ; Kandoth C, Schultz N, Cherniack AD, Akbani R, Liu Y, Shen H, Robertson AG, Pashtan I, Shen R, et al: Integrated genomic characterization of endometrial carcinoma. Nature. 497:67–73. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Talhouk A, McConechy MK, Leung S, Li-Chang HH, Kwon JS, Melnyk N, Yang W, Senz J, Boyd N, Karnezis AN, et al: A clinically applicable molecular-based classification for endometrial cancers. Br J Cancer. 113:299–310. 2015. View Article : Google Scholar : PubMed/NCBI

14 

ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature. 489:57–74. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Wilusz JE, Sunwoo H and Spector DL: Long noncoding RNAs: Functional surprises from the RNA world. Genes Dev. 23:1494–1504. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Gibb EA, Vucic EA, Enfield KS, Stewart GL, Lonergan KM, Kennett JY, Becker-Santos DD, MacAulay CE, Lam S, Brown CJ, et al: Human cancer long non-coding RNA transcriptomes. PLoS One. 6:e259152011. View Article : Google Scholar : PubMed/NCBI

17 

Gibb EA, Brown CJ and Lam WL: The functional role of long non-coding RNA in human carcinomas. Mol Cancer. 10:382011. View Article : Google Scholar : PubMed/NCBI

18 

Angrand PO, Vennin C, Le Bourhis X and Adriaenssens E: The role of long non-coding RNAs in genome formatting and expression. Front Genet. 6:1652015. View Article : Google Scholar : PubMed/NCBI

19 

Esteller M: Non-coding RNAs in human disease. Nat Rev Genet. 12:861–874. 2011. View Article : Google Scholar : PubMed/NCBI

20 

St Laurent G, Wahlestedt C and Kapranov P: The landscape of long noncoding RNA classification. Trends Genet. 31:239–251. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Ravo M, Cordella A, Rinaldi A, Bruno G, Alexandrova E, Saggese P, Nassa G, Giurato G, Tarallo R, Marchese G, et al: Small non-coding RNA deregulation in endometrial carcinogenesis. Oncotarget. 6:4677–4691. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Nassa G, Giurato G, Cimmino G, Rizzo F, Ravo M, Salvati A, Nyman TA, Zhu Y, Vesterlund M, Lehtiö J, et al: Splicing of platelet resident pre-mRNAs upon activation by physiological stimuli results in functionally relevant proteome modifications. Sci Rep. 8:4982018. View Article : Google Scholar : PubMed/NCBI

23 

Tarallo R, Giurato G, Bruno G, Ravo M, Rizzo F, Salvati A, Ricciardi L, Marchese G, Cordella A, Rocco T, et al: The nuclear receptor ERbeta engages AGO2 in regulation of gene transcription, RNA splicing and RISC loading. Genome Biol. 18:1892017. View Article : Google Scholar : PubMed/NCBI

24 

Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, Batut P, Chaisson M and Gingeras TR: STAR: Ultrafast universal RNA-seq aligner. Bioinformatics. 29:15–21. 2013. View Article : Google Scholar : PubMed/NCBI

25 

Anders S, Pyl PT and Huber W: HTSeq-a Python framework to work with high-throughput sequencing data. Bioinformatics. 31:166–169. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Love MI, Huber W and Anders S: Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15:5502014. View Article : Google Scholar : PubMed/NCBI

27 

Gaujoux R and Seoighe C: A flexible R package for nonnegative matrix factorization. BMC Bioinformatics. 11:3672010. View Article : Google Scholar : PubMed/NCBI

28 

Dweep H and Gretz N: miRWalk2.0: A comprehensive atlas of microRNA-target interactions. Nat Methods. 12:6972015. View Article : Google Scholar : PubMed/NCBI

29 

Wang W, Zhuang Q, Ji K, Wen B, Lin P, Zhao Y, Li W and Yan C: Identification of miRNA, lncRNA and mRNA-associated ceRNA networks and potential biomarker for MELAS with mitochondrial DNA A3243G mutation. Sci Rep. 7:416392017. View Article : Google Scholar : PubMed/NCBI

30 

Zhao Z, Bai J, Wu A, Wang Y, Zhang J, Wang Z, Li Y, Xu J and Li X: Co-LncRNA: Investigating the lncRNA combinatorial effects in GO annotations and KEGG pathways based on human RNA-Seq data. Database. 2015:bav0822015. View Article : Google Scholar : PubMed/NCBI

31 

Derrien T, Johnson R, Bussotti G, Tanzer A, Djebali S, Tilgner H, Guernec G, Martin D, Merkel A, Knowles DG, et al: The GENCODE v7 catalog of human long noncoding RNAs: Analysis of their gene structure, evolution, and expression. Genome Res. 22:1775–1789. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Talhouk A and McAlpine JN: New classification of endometrial cancers: The development and potential applications of genomic-based classification in research and clinical care. Gynecol Oncol Res Pract. 3:142016. View Article : Google Scholar : PubMed/NCBI

33 

Vallone C, Rigon G, Gulia C, Baffa A, Votino R, Morosetti G, Zaami S, Briganti V, Catania F, Gaffi M, et al: Non-coding RNAs and endometrial cancer. Genes. 9:E1872018. View Article : Google Scholar : PubMed/NCBI

34 

Xu J, Qian Y, Ye M, Fu Z, Jia X, Li W, Xu P, Lv M, Huang L, Wang L, et al: Distinct expression profile of lncRNA in endometrial carcinoma. Oncol Rep. 36:3405–3412. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Fatima R, Akhade VS, Pal D and Rao SM: Long noncoding RNAs in development and cancer: Potential biomarkers and therapeutic targets. Mol Cell Ther. 3:52015. View Article : Google Scholar : PubMed/NCBI

36 

Schmitt AM and Chang HY: Long noncoding RNAs in cancer pathways. Cancer Cell. 29:452–463. 2016. View Article : Google Scholar : PubMed/NCBI

37 

Chen BJ, Byrne FL, Takenaka K, Modesitt SC, Olzomer EM, Mills JD, Farrell R, Hoehn KL and Janitz M: Transcriptome landscape of long intergenic non-coding RNAs in endometrial cancer. Gynecol Oncol. 147:654–662. 2017. View Article : Google Scholar : PubMed/NCBI

38 

Huang J, Ke P, Guo L, Wang W, Tan H, Liang Y and Yao S: Lentivirus-mediated RNA interference targeting the long noncoding RNA HOTAIR inhibits proliferation and invasion of endometrial carcinoma cells in vitro and in vivo. Int J Gynecol Cancer. 24:635–642. 2014. View Article : Google Scholar : PubMed/NCBI

39 

Shang C, Lang B, Ao CN and Meng L: Long non-coding RNA tumor suppressor candidate 7 advances chemotherapy sensitivity of endometrial carcinoma through targeted silencing of miR-23b. Tumour Biol. 39:10104283177078832017. View Article : Google Scholar : PubMed/NCBI

40 

Sun KX, Wu DD, Chen S, Zhao Y and Zong ZH: LncRNA MEG3 inhibit endometrial carcinoma tumorigenesis and progression through PI3K pathway. Apoptosis. 22:1543–1552. 2017. View Article : Google Scholar : PubMed/NCBI

41 

Wang D, Wang D, Wang N, Long Z and Ren X: Long non-coding RNA BANCR promotes endometrial cancer cell proliferation and invasion by regulating MMP2 and MMP1 via ERK/MAPK signaling pathway. Cell Physiol Biochem. 40:644–656. 2016. View Article : Google Scholar : PubMed/NCBI

42 

Yang L, Zhang J, Jiang A, Liu Q, Li C, Yang C and Xiu J: Expression profile of long non-coding RNAs is altered in endometrial cancer. Int J Clin Exp Med. 8:5010–5021. 2015.PubMed/NCBI

43 

Xin W, Liu X, Ding J, Zhao J, Zhou Y, Wu Q and Hua K: Long non-coding RNA derived miR-205-5p modulates human endometrial cancer by targeting PTEN. Am J Transl Res. 7:2433–2441. 2015.PubMed/NCBI

44 

Zhai W, Li X, Wu S, Zhang Y, Pang H and Chen W: Microarray expression profile of lncRNAs and the upregulated ASLNC04080 lncRNA in human endometrial carcinoma. Int J Oncol. 46:2125–2137. 2015. View Article : Google Scholar : PubMed/NCBI

45 

Salmena L, Poliseno L, Tay Y, Kats L and Pandolfi PP: A ceRNA hypothesis: The Rosetta Stone of a hidden RNA language? Cell. 146:353–358. 2011. View Article : Google Scholar : PubMed/NCBI

46 

Tay Y, Rinn J and Pandolfi PP: The multilayered complexity of ceRNA crosstalk and competition. Nature. 505:344–352. 2014. View Article : Google Scholar : PubMed/NCBI

47 

Cheng DL, Xiang YY, Ji LJ and Lu XJ: Competing endogenous RNA interplay in cancer: Mechanism, methodology, and perspectives. Tumour Biol. 36:479–488. 2015. View Article : Google Scholar : PubMed/NCBI

48 

Lu M, Xu X, Xi B, Dai Q, Li C, Su L, Zhou X, Tang M, Yao Y and Yang J: Molecular network-based identification of competing endogenous RNAs in thyroid carcinoma. Genes. 9:E442018. View Article : Google Scholar : PubMed/NCBI

49 

Liu C, Zhang YH, Deng Q, Li Y, Huang T, Zhou S and Cai YD: Cancer-related triplets of mRNA-lncRNA-miRNA revealed by integrative network in uterine corpus endometrial carcinoma. Biomed Res Int. 2017:38595822017.PubMed/NCBI

50 

Jia H, Zhang Z, Zou D, Wang B, Yan Y, Luo M, Dong L, Yin H, Gong B, Li Z, et al: MicroRNA-10a is down-regulated by DNA methylation and functions as a tumor suppressor in gastric cancer cells. PLoS One. 9:e880572014. View Article : Google Scholar : PubMed/NCBI

51 

Zou D, Zhou Q, Wang D, Guan L, Yuan L and Li S: The downregulation of MicroRNA-10b and its role in cervical cancer. Oncol Res. 24:99–108. 2016. View Article : Google Scholar : PubMed/NCBI

52 

Snowdon J, Zhang X, Childs T, Tron VA and Feilotter H: The microRNA-200 family is upregulated in endometrial carcinoma. PLoS One. 6:e228282011. View Article : Google Scholar : PubMed/NCBI

53 

Panda H, Pelakh L, Chuang TD, Luo X, Bukulmez O and Chegini N: Endometrial miR-200c is altered during transformation into cancerous states and targets the expression of ZEBsVEGFAFLT1IKKbetaKLF9, and FBLN5. Reprod Sci. 19:786–796. 2012. View Article : Google Scholar : PubMed/NCBI

54 

Korani M, Fallah S, Tehranian A, Nourbakhsh M, Samadikuchaksaraei A, Pour MS and Maleki J: The evaluation of the FOXO1, KLF9 and YT521 genes expression in human endometrial cancer. Clin Lab. 59:483–489. 2013. View Article : Google Scholar : PubMed/NCBI

55 

Simmons CD, Pabona JM, Heard ME, Friedman TM, Spataro MT, Godley AL, Simmen FA, Burnett AF and Simmen RC: Krüppel-like factor 9 loss-of-expression in human endometrial carcinoma links altered expression of growth-regulatory genes with aberrant proliferative response to estrogen. Biol Reprod. 85:378–385. 2011. View Article : Google Scholar : PubMed/NCBI

56 

Liu Y, Nan F, Lu K and Wang Y, Liu Y, Wei S, Wu R and Wang Y: Identification of key genes in endometrioid endometrial adenocarcinoma via TCGA database. Cancer Biomark. 21:11–21. 2017. View Article : Google Scholar : PubMed/NCBI

57 

Honkavuori-Toivola M, Santala M, Soini Y, Turpeenniemi- Hujanen T and Talvensaari-Mattila A: Combination of strong MMP-2 and weak TIMP-2 immunostainings is a significant prognostic factor in endometrial carcinoma. Dis Markers. 35:261–266. 2013. View Article : Google Scholar : PubMed/NCBI

58 

Dai Y, Xia W, Song T, Su X, Li J, Li S, Chen Y, Wang W, Ding H, Liu X, et al: MicroRNA-200b is overexpressed in endometrial adenocarcinomas and enhances MMP2 activity by downregulating TIMP2 in human endometrial cancer cell line HEC-1A cells. Nucleic Acid Ther. 23:29–34. 2013. View Article : Google Scholar : PubMed/NCBI

59 

Liu C, Yang Z, Deng Z, Zhou Y, Gong Q, Zhao R and Chen T: Upregulated lncRNA ADAMTS9-AS2 suppresses progression of lung cancer through inhibition of miR-223-3p and promotion of TGFBR3. IUBMB Life. 70:536–546. 2018. View Article : Google Scholar : PubMed/NCBI

60 

Yao J, Zhou B, Zhang J, Geng P, Liu K, Zhu Y and Zhu W: A new tumor suppressor LncRNA ADAMTS9-AS2 is regulated by DNMT1 and inhibits migration of glioma cells. Tumour Biol. 35:7935–7944. 2014. View Article : Google Scholar : PubMed/NCBI

61 

Guo Q, Qian Z, Yan D, Li L and Huang L: LncRNA-MEG3 inhibits cell proliferation of endometrial carcinoma by repressing Notch signaling. Biomed Pharmacother. 82:589–594. 2016. View Article : Google Scholar : PubMed/NCBI

62 

Terashima M, Tange S, Ishimura A and Suzuki T: MEG3 long noncoding RNA contributes to the epigenetic regulation of epithelial-mesenchymal transition in lung cancer cell lines. J Biol Chem. 292:82–99. 2017. View Article : Google Scholar : PubMed/NCBI

63 

Yang X, Wang CC, Lee WYW, Trovik J, Chung TKH and Kwong J: Long non-coding RNA HAND2-AS1 inhibits invasion and metastasis in endometrioid endometrial carcinoma through inactivating neuromedin U. Cancer Lett. 413:23–34. 2018. View Article : Google Scholar : PubMed/NCBI

64 

Wang H, Niu L, Jiang S, Zhai J, Wang P, Kong F and Jin X: Comprehensive analysis of aberrantly expressed profiles of lncRNAs and miRNAs with associated ceRNA network in muscle-invasive bladder cancer. Oncotarget. 7:86174–86185. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2019
Volume 41 Issue 2

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ravo M, Cordella A, Saggese P, Rinaldi A, Castaldi MA, Nassa G, Giurato G, Zullo F, Weisz A, Tarallo R, Tarallo R, et al: Identification of long non‑coding RNA expression patterns useful for molecular‑based classification of type I endometrial cancers. Oncol Rep 41: 1209-1217, 2019
APA
Ravo, M., Cordella, A., Saggese, P., Rinaldi, A., Castaldi, M.A., Nassa, G. ... Guida, M. (2019). Identification of long non‑coding RNA expression patterns useful for molecular‑based classification of type I endometrial cancers. Oncology Reports, 41, 1209-1217. https://doi.org/10.3892/or.2018.6880
MLA
Ravo, M., Cordella, A., Saggese, P., Rinaldi, A., Castaldi, M. A., Nassa, G., Giurato, G., Zullo, F., Weisz, A., Tarallo, R., Rizzo, F., Guida, M."Identification of long non‑coding RNA expression patterns useful for molecular‑based classification of type I endometrial cancers". Oncology Reports 41.2 (2019): 1209-1217.
Chicago
Ravo, M., Cordella, A., Saggese, P., Rinaldi, A., Castaldi, M. A., Nassa, G., Giurato, G., Zullo, F., Weisz, A., Tarallo, R., Rizzo, F., Guida, M."Identification of long non‑coding RNA expression patterns useful for molecular‑based classification of type I endometrial cancers". Oncology Reports 41, no. 2 (2019): 1209-1217. https://doi.org/10.3892/or.2018.6880