Open Access

Bioinformatics analysis of the expression of HOXC13 and its role in the prognosis of breast cancer

  • Authors:
    • Changyou Li
    • Junwei Cui
    • Li Zou
    • Lizhang Zhu
    • Wei Wei
  • View Affiliations

  • Published online on: November 22, 2019     https://doi.org/10.3892/ol.2019.11140
  • Pages: 899-907
  • Copyright: © Li et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The homeobox (HOX) genes, a class of transcription factors, are known to promote embryonic development and induce tumor formation. To date, the HOXA and HOXB gene families have been reported to be associated with breast cancer. However, the expression and exact role of homeobox C13 (HOXC13) in breast cancer has not yet been investigated. In the present study, the HOXC13 expression in human breast cancer was evaluated using the Oncomine database and Cancer Cell Line Encyclopedia (CCLE). Next, the Gene expression‑based Outcome for Breast cancer online database, cBioportal, University of California Santa Cruz Xena browser and bc‑GenExMinerv were used to explore the specific expression of HOXC13 in breast cancer. The methylation and mutation status of HOXC13 in breast cancer was then validated using the CCLE and cBioportal databases. Finally, the co‑expression of HOX transcript antisense RNA (HOTAIR) and HOXC13 in breast cancer were analyzed and their impact on clinical prognosis determined. It was found that the expression of HOXC13 was high in breast cancer compared with other types of cancer, such as gastric cancer and colon cancer. Following co‑­expression analysis, a significant positive association was identified between HOTAIR and HOXC13. An association between HOTAIR and HOXC13, and lymph node and distant metastasis recurrence was also revealed during the development of breast cancer. Of note, survival analysis showed that high expression of HOTAIR and HOXC13 predicted poor prognosis. These findings revealed that HOXC13 plays an important role in the progression of breast cancer. However, the specific mechanism needs to be confirmed by subsequent experiments.

Introduction

Breast cancer, the most common type of cancer in women, presents a challenge for global research (1). Molecular research on breast cancer was at a bottleneck, until the identification of a correlation between BRCA1/2 DNA repair associated and breast cancer was reported and marked great progress in the research, treatment and prognosis of breast cancer (2,3). Following this, genetic susceptibility has become the focus of breast cancer research (4). However, in the context of big data, with the development of high-throughput sequencing technology, it is not difficult to find susceptibility genes.

The Homeobox (HOX) genes is a large class of transcription factors that play an important role in embryogenesis and oncogenesis, as well as the distribution of fat and hair in body (57). In humans, the HOX gene family contains 39 HOX genes located on 4 different chromosomes (7p15, 17q21.2, 12q13 and 2q31) (8). The 39 HOX genes are divided into 4 clusters (HOXA, HOXB, HOXC and HOXD) (9). Each HOX gene contains a well-conserved DNA sequence, known as the homeobox (10). The unique expression pattern, including mutation, and dependent mechanism of the HOX genes regulates, to some extent, the embryonic development of vertebrates (11,12). When HOX protein expression is upregulated, it may lead to cancer (5). It has also been reported that the HOXC gene family is highly expressed in certain solid tumors, including lung, prostate and colon cancer (13,14). The HOXA and HOXB gene families have a similar expression in breast cancer, which is derived from the ectoderm. Whether the expression level of the HOX gene follows the origin of the germ layer in cancer requires further investigation. A study has reported that HOXC13, a member of the HOXC gene family, is highly expressed in the MCF-7 cell line (15). Thus, the present study aimed to explore the expression and significance of HOXC13 in breast cancer.

In the present study, the Oncomine and tumor public databases (bc-GenExMinerv 4.2; GOBO database; CCLE) were used to analyze the expression level of HOXC13 in different types of cancer including breast cancer. HOXC13 was then further investigated in breast cancer. The expression and co-expression of HOXC13 in breast cancer was re-analyzed using the University of California, Santa Cruz (UCSC) cancer genomics browser. Finally, the clinical significance of HOXC13 in breast cancer was further explored.

Materials and methods

Oncomine database verification

The Oncomine (www.oncomine.org) database is a public bioinformatics database containing gene expression data set that has become an industry-standard tool cited in >1,100 peer-reviewed journal articles (16,17). The Oncomine platform has been used as a foundation for ground-breaking discoveries with unique features that include scalability, high quality, consistency and standardized analysis (18). In order to screen out the most meaningful RNA probes, the paired Student's t-test was used to generate P-values to compare expression differences between cancer and healthy adjacent non-cancerous tissues. Relevant statistical indicators were used as follows: P<1×10−4, fold change >4 and gene ranking in the top 10%. Moreover, the Oncomine database was used to explore the co-expression analysis of HOXC13 in breast cancer. The search term ‘HOXC13’ was used, followed by coexpression analysis and selecting the cancer type as breast cancer. Lastly, the database of TCGA breast was chosen. Furthermore, the same cut-off values used as aforementioned.

Cancer cell line encyclopedia (CCLE) verification

The CCLE (portals.broadinstitute.org) provides public access to genomic data, analysis and visualization for >1,100 cell lines from various tumors, such as gastric cancer cell line AGS and intestinal cancer cell line SW480 (19,20). Each gene of the human genome has multiple datasets and data identifiers, obtained by high-throughput sequencing. The 5 major dataset types are copy number, mRNA expression (Affymetrix), reverse phase protein array, reduced representation bisulfite sequencing, and mRNA expression (RNA sequencing). The expression and methylation level of HOXC13 was analyzed in each tumor cell line using CCLE, using the search term ‘HOXC13’.

Gene expression-based Outcome for Breast cancer Online (GOBO) analysis

The GOBO database (version 1.0.3; co.bmc.lu.se/gobo/gsa_cellines.pl) is a user-friendly public database. It allows for rapid assessment of gene expression levels, identification of co-expressed genes and association with outcome for single genes, gene sets or gene signatures in an 1,881-sample breast cancer dataset (21). The most important functionality of the GOBO database is the possibility of investigating gene expression levels in breast cancer subgroups and cell lines for gene sets (22). Breast cancer subtypes are classified into basal A, basal B and luminal in the GOBO database. A correlation map is a square table where each line and column represent a gene. Each cell represents an interaction between two genes and is colored according to the value of the Pearson's correlation coefficient between these two genes, from dark blue (coefficient=−1) to dark red (coefficient=1). Cells from the diagonal of the correlation map represent the interaction of a gene with itself and are colored black.

UCSC cancer genomics browser analysis

UCSC (xena.ucsc.edu) is an online exploration tool for public and private multi-omics functional genomics and clinical/phenotype data (23). The Cancer Genome Atlas (TCGA; http://www.cancer.gov/about-nci/organization/ccg/research/structural-genomics/tcga) were used in the present study. Using the UCSC database, a heat map of HOXC13 expression in various subtypes of breast cancer, such as LuminalA, luminalB, normal-like, Basal-like and Her2-riched was generated. At the same time, the co-expression of HOXC13 in breast cancer was analyzed.

cBioPortal database analysis

The public cBioPortal site (www.cbioportal.org; last entered, 28th March 2019) is hosted by the Center for Molecular Oncology at Memorial Sloan Kettering Cancer Center. The cBioportal database has been recognized as a way to verify gene mutations (2426). This database can also analyze gene expression in different tumors and different studies (23,27). This database has been recognized by multiple studies (26,28). The online cBioPortal for Cancer Genomics was used to provide mutations of HOXC13 (including nonstart, missense, truncation and missense) and its expression in different studies (28). The cancer types and databases used were breast (TCGA2015), breast (TCGA), breast invasive carcinoma breast (TCGA PanCan) (2934).

Breast cancer gene-expression miner (bc-GenExMiner) analysis

bc-GenExMinerv (version 4.2) is a statistical mining tool of published annotated genomic data (35,36). The statistical analyses are grouped in three modules: Expression, prognosis and correlation. The co-expression of HOXC13 in breast cancer was explored using this database; each study is validated across multiple databases to avoid discrepancies in individual databases. At the same time, the effects of HOXC13 and HOX transcript antisense RNA (HOTAIR) on the survival prognosis in breast cancer were also analyzed.

Statistical analysis

Pearson test and Spearman's rank test were used to evaluate coexpression. The analysis criteria selected were as follows: Gene, nodal and estrogen receptor status of the cohorts to be explored, event on which survival analysis will be based and splitting criterion of median HOXC13 expression. To analyze the prognostic value of HOXC13 and HOTAIR, the patient samples are split into two groups according to the median expressions of HOXC13 and HOTAIR. The two patient cohorts were compared by a Kaplan-Meier survival plot, and the hazard ratio with 95% confidence intervals and logrank P-value are calculated. Significance was determined by the P-value provided by each database.

Results

HOXC13 mRNA expression in human cancer

HOXC13 has been proven to be highly expressed in digestive tract-derived tumors (3739). However, to the best of our knowledge, there have been no reports on the expression of HOXC13 in breast cancer. Therefore the expression levels of HOXC13 in various human tumors from the Oncomine database and CCLE were determined. A simultaneous fold change of >4, gene rank of >10% and P<1×10−4 was set as the threshold. To our surprise, HOXC13 was found by high-throughput sequencing and biological gene chip technology to be abnormally highly expressed in breast cancer (Fig. 1A). Only breast cancer revealed significant unique analysis in the Oncomine database when the fold change was >4.

HOXC13 mRNA expression in breast cancer

The high expression of HOXC13 in breast cancer has been preliminary confirmed in the Oncomine database, GOBO database and CCLE. From the CCLE database, it was found that at the RNAseq level, the expression level of HOXC13 in breast cancer ranked fourth and ranked first in the Affy level. However, there is no report on the specific expression of HOXC13 in breast cancer. Therefore, its specific expression in various subtypes of breast cancer was further explored. HOXC13 was analyzed in various tumor types via Oncomine database and GOBO database and explored HOXC13 in various breast cancer cells via GOBO database and CCLE. HOXC13 was found to be highly expressed in invasive and luminal-like breast cancer than in any other subtype (Figs. 2 and 3). Such expression characteristics were consistent with breast cancer cell lines and tissues (Figs. 2D and 3). Using the UCSC cancer genomics browser analysis, the heat map of the gene and exon expression of HOXC13 in various subtypes of breast cancer was obtained (Fig. 4A). Furthermore, the expression of HOXC13 in the different data sets, including breast (TCGA 2015), breast (TCGA), breast invasive carcinoma breast (TCGA PanCan), was explored using the cBioPortal database. The expression characteristics of breast cancer in multiple data sets such as amplification and missense mutations, were demonstrated (Fig. 4B).

HOXC13 methylation and mutation in human breast cancer

The bubble chart shows the methylation level of HOXC13 in breast cancer cell lines from the CCLE (Fig. 5A). HOXC13 is highly methylated at three sites (positions 54,330,731, 54,330,950 and 54,330,957) on chromosome 12 from methylation and coverage (Fig. 5A). The discovery of cpG island methylation further supported the high expression of HOXC13 in breast cancer. cBioPortal was used to assess the frequency of HOXC13 mutations in breast cancer (Figs. 4B and 5). HOXC13 contains multiple mutations in breast cancer such as amplification, gain, missense and truncation (Fig. 4B). Missense and truncation are two major forms of mutation (Fig. 5).

Co-expression of HOXC13 mRNA in breast cancer

To investigate the reason for the high expression of HOXC13 in breast cancer, bc-GenExMiner version 4.2 was used to analyze the potential co-expression of HOXC13 in breast cancer. It was found that HOTAIR and HOXC13 are highly co-expressed in breast cancer (Fig. 6A). Furthermore, to verify the co-expression of HOXC13 and HOTAIR in breast cancer, their co-expression heat maps were obtained and mined (Fig. 6B) and correlation analysis was performed using the UCSC Xena browser (Fig. 6C).

Impact of HOXC13 and HOTAIR on the prognosis of patients with breast cancer

To verify the impact of the high expression of HOXC13 and HOTAIR on patients with breast cancer, prognostic analysis of HOXC13 and HOTAIR in breast cancer was performed and HOXC13 and HOTAIR were found to have a negative impact on the prognosis of patients with tumor and lymph node metastasis (Fig. 7).

Discussion

The mammary gland is an appendage of the ectoderm, whose formation begins during embryonic development (40). Moreover, breast cancer cells remain highly associated with ectoderm cells (41). Studies have shown that the HOX gene plays an important role in embryonic development and tumor formation (9,4244). However, to the best of our knowledge, no research has reported the exact role of HOXC13 in breast cancer so far.

The HOXC13 gene is considered an element of hair morphogenesis, and the HOXC13 protein is a member of the human replication complex in growing cells (45,46). This id the same for breast adenocarcinoma MCF7 lines (46). HOXC13 can promote the expression of a series of proto-oncogenes, including topoisomerase I and II, and allow these expression products to form a replication complex (47,48). Therefore, HOXC13 is involved in the development of tumors. In addition, HOXC13 increases the metastasis of ectodermal-derived melanoma (49). HOTAIR is the product of the transcription of the HOXC gene cluster antisense strand (50). HOTAIR is transcribed from the mammalian HOXC gene cluster on chromosome 12q13.13 (51,52). HOTAIR predicts poor prognosis in tumor cell cycle and metastasis (53). It has been reported that HOTAIR upregulates HOX in colon cancer (39). A strong co-expression of HOTAIR and HOXC13 was confirmed in proximal and distal colon cancer, suggesting that HOTAIR and HOXC13 could promote tumor and lymph node metastasis (39,5456).

In the present study, it was first identified that HOXC13 is highly expressed in breast cancer both at the cellular and tissue levels. This was a finding from the co-verification of the data from TCGA's Oncomine or GSE's bc-GenExMiner. It was found for the first time that HOXC13 is most highly expressed in luminal-like subtype of breast cancer. This laid the foundation for the future study of the relationship between HOXC13 and breast cancer surface hormone receptors (estrogen receptor and progesterone receptor). In order to explore the high expression of HOXC13 in breast cancer, its methylation and mutation status were investigated. To our surprise, three sites on chromosome 12 were found to be consistently highly methylated in different breast cancer cell lines. Certain studies have reported that HOX gene methylation regulates hereditary breast cancer (5759). In addition, the methylation level of HOXA11 is significantly higher in breast cancer compared with that in normal tissues, and is positively associated with family history and lymph node metastasis in breast cancer (59). Furthermore, the methylation level of HOXD13 in breast cancer is almost identical to that of HOXA11, and leads to shorter survival time (58). Therefore, the present findings and the HOX gene family have consistent trends in methylation levels and similar prognostic effects in breast cancer. However, further studies on the specific regulation mechanism of HOXC13 methylation in HOXC13 transcription is required. The present study is the first to report the mutation of HOXC13 in breast cancer. Missense may be an indispensable cause of the high expression of HOXC13 in breast cancer.

The lncRNA HOTAIR is derived from the region between HOXC11 and HOXC12 (51). It has been shown that HOXC10, HOXC11, HOXC12 and HOXC13 are adjacent to each other in the HOXC gene cluster (52). Simultaneously, the HOXC distal enhancer has non-specific enhancement of HOXC10 and HOTAIR enhancer activity, promoting the HOXC10 and HOTAIR expression (60). On the other hand, specific intergenic non-coding RNAs (including HOTAIR) in the HOX loci can directly modulate the expression of the HOX gene in normal and cancer status (61). This has been confirmed in colon cancer (39,55). Therefore the mechanism between HOXC13 and HOTAIR will be explored further in this respect. In the present study, the co-expression of HOTAIR and HOXC13 provided a new direction for studying the function of HOTAIR in breast cancer. In addition, a study has identified through meta-analysis that HOTAIR has a statistically significant effect on lymph node and distant metastasis in various types of cancer, including breast cancer, gastric cancer and colorectal cancer, which was consistent with our conclusion (62). The present data showed that HOTAIR and HOXC13 were significantly associated with lymph node metastasis and distant metastasis recurrence. Furthermore, they were shown to have a significant impact on survival period.

In conclusion, the present study was performed using public databases and revealed the expression and clinical significance of HOXC13 in breast cancer. However, the specific interactions and mechanisms involved require further experimental verification, which will be performed in future studies.

Acknowledgements

Not applicable.

Funding

This study was supported by the Shenzhen Science and Research Innovation Foundation (grant no. JCYJ20170815090309586).

Availability of data and materials

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

Authors' contributions

WW contributed to the experimental design and fundraising. JWC and LZ contributed to the acquisition of data. LZZ assisted in the data processing. CL processed the data and wrote the 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.

References

1 

Barrios CH, Reinert T and Werutsky G: Global breast cancer research: Moving forward. Am Soc Clin Oncol Educ Book. 38:441–450. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Harbeck N and Gnant M: Breast cancer. Lancet. 389:1134–1150. 2017. View Article : Google Scholar : PubMed/NCBI

3 

Nathanson KL and Domchek SM: Therapeutic approaches for women predisposed to breast cancer. Annu Rev Med. 62:295–306. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Rousset-Jablonski C and Gompel A: Screening for familial cancer risk: Focus on breast cancer. Maturitas. 105:69–77. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Luo Z, Rhie SK and Farnham PJ: The enigmatic HOX genes: Can we crack their code? Cancers (Basel). 11(pii): E3232019. View Article : Google Scholar : PubMed/NCBI

6 

Turcotte M, Abadi A, Peralta-Romero J, Suarez F, Reddon H, Gomez-Zamudio J, Burguete-Garcia AI, Cruz M and Meyre D: Genetic contribution to waist-to-hip ratio in Mexican children and adolescents based on 12 loci validated in European adults. Int J Obes (Lond). 43:13–22. 2019. View Article : Google Scholar : PubMed/NCBI

7 

Liu F, Chen Y, Zhu G, Hysi PG, Wu S, Adhikari K, Breslin K, Pospiech E, Hamer MA, Peng F, et al: Meta-analysis of genome-wide association studies identifies 8 novel loci involved in shape variation of human head hair. Hum Mol Genet. 27:559–575. 2018. View Article : Google Scholar : PubMed/NCBI

8 

Li L, Wang Y, Song G, Zhang X, Gao S and Liu H: HOX cluster-embedded antisense long non-coding RNAs in lung cancer. Cancer Lett. 450:14–21. 2019. View Article : Google Scholar : PubMed/NCBI

9 

Bhatlekar S, Fields JZ and Boman BM: Role of HOX genes in stem cell differentiation and cancer. Stem Cells Int. 2018:35694932018. View Article : Google Scholar : PubMed/NCBI

10 

Vaquerizas JM, Kummerfeld SK, Teichmann SA and Luscombe NM: A census of human transcription factors: Function, expression and evolution. Nat Rev Genet. 10:252–263. 2009. View Article : Google Scholar : PubMed/NCBI

11 

Gordon J: Hox genes in the pharyngeal region: How Hoxa3 controls early embryonic development of the pharyngeal organs. Int J Dev Biol. 62:775–783. 2018. View Article : Google Scholar : PubMed/NCBI

12 

Mallo M: The vertebrate tail: A gene playground for evolution. Cell Mol Life Sci. Sep 26–2019.(Epub ahead of print). View Article : Google Scholar : PubMed/NCBI

13 

Bhatlekar S, Fields JZ and Boman BM: HOX genes and their role in the development of human cancers. J Mol Med (Berl). 92:811–823. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Yao Y, Luo J, Sun Q, Xu T, Sun S, Chen M, Lin X, Qian Q, Zhang Y, Cao L, et al: HOXC13 promotes proliferation of lung adenocarcinoma via modulation of CCND1 and CCNE1. Am J Cancer Res. 7:1820–1834. 2017.PubMed/NCBI

15 

Svingen T and Tonissen KF: Altered HOX gene expression in human skin and breast cancer cells. Cancer Biol Ther. 2:518–523. 2003. View Article : Google Scholar : PubMed/NCBI

16 

Shi S and Zhang ZG: Role of Sp1 expression in gastric cancer: A meta-analysis and bioinformatics analysis. Oncol Lett. 18:4126–4135. 2019.PubMed/NCBI

17 

Cheng L, Shi L and Dai H: Bioinformatics prognostic biomarkers among Krüppel-like transcription factors (KLFs) in breast cancer. Cancer Biomark. Oct 12–2019.(Epub ahead of print). View Article : Google Scholar

18 

Yang K, Gao J and Luo M: Identification of key pathways and hub genes in basal-like breast cancer using bioinformatics analysis. Onco Targets Ther. 12:1319–1331. 2019. View Article : Google Scholar : PubMed/NCBI

19 

Cheng L, Pandya PH, Liu E, Chandra P, Wang L, Murray ME, Carter J, Ferguson M, Saadatzadeh MR, Bijangi-Visheshsaraei K, et al: Integration of genomic copy number variations and chemotherapy-response biomarkers in pediatric sarcoma. BMC Med Genomics. 12 (Suppl 1):S232019. View Article : Google Scholar

20 

Streit M, Gratzl S, Stitz H, Wernitznig A, Zichner T and Haslinger C: Ordino: A visual cancer analysis tool for ranking and exploring genes, cell lines and tissue samples. Bioinformatics. 35:3140–3142. 2019. View Article : Google Scholar : PubMed/NCBI

21 

Fernández-Nogueira P, Bragado P, Almendro V, Ametller E, Rios J, Choudhury S, Mancino M and Gascón P: Differential expression of neurogenes among breast cancer subtypes identifies high risk patients. Oncotarget. 7:5313–5326. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Zen g, Xiao Y, Zhu J, Peng C, Liang W and Lin H: Knockdown of nucleophosmin 1 suppresses proliferation of triple-negative breast cancer cells through activating CDH1/Skp2/p27kip1 pathway. Cancer Manag Res. 11:143–156. 2018. View Article : Google Scholar : PubMed/NCBI

23 

Klonowska K, Czubak K, Wojciechowska M, Handschuh L, Zmienko A, Figlerowicz M, Dams-Kozlowska H and Kozlowski P: Oncogenomic portals for the visualization and analysis of genome-wide cancer data. Oncotarget. 7:176–192. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Wen W, Gong J, Wu P, Zhao M, Wang M, Chen H and Sun J: Mutations in gliclazide-associated genes may predict poor bladder cancer prognosis. FEBS Open Bio. 9:457–467. 2019.PubMed/NCBI

25 

Chen E, Qin X, Peng K, Xu X, Li W, Cheng X, Tang C, Cui Y, Wang Z and Liu T: Identification of potential therapeutic targets among CXC chemokines in breast tumor microenvironment using integrative bioinformatics analysis. Cell Physiol Biochem. 45:1731–1746. 2018. View Article : Google Scholar : PubMed/NCBI

26 

Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, Jacobsen A, Byrne CJ, Heuer ML, Larsson E, et al: The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2:401–404. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Kline CLB, Ralff MD, Lulla AR, Wagner JM, Abbosh PH, Dicker DT, Allen JE and El-Deiry WS: Role of dopamine receptors in the anticancer activity of ONC201. Neoplasia. 20:80–91. 2018. View Article : Google Scholar : PubMed/NCBI

28 

Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, Sun Y, Jacobsen A, Sinha R, Larsson E, et al: Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 6:pl12013. View Article : Google Scholar : PubMed/NCBI

29 

Banerji S, Cibulskis K, Rangel-Escareno C, Brown KK, Carter SL, Frederick AM, Lawrence MS, Sivachenko AY, Sougnez C, Zou L, et al: Sequence analysis of mutations and translocations across breast cancer subtypes. Nature. 486:405–409. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Eirew P, Steif A, Khattra J, Ha G, Yap D, Farahani H, Gelmon K, Chia S, Mar C, Wan A, et al: Dynamics of genomic clones in breast cancer patient xenografts at single-cell resolution. Nature. 518:422–426. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Martelotto LG, De Filippo MR, Ng CK, Natrajan R, Fuhrmann L, Cyrta J, Piscuoglio S, Wen HC, Lim RS, Shen R, et al: Genomic landscape of adenoid cystic carcinoma of the breast. J Pathol. 237:179–189. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Razavi P, Chang MT, Xu G, Bandlamudi C, Ross DS, Vasan N, Cai Y, Bielski CM, Donoghue MTA, Jonsson P, et al: The genomic landscape of endocrine-resistant advanced breast cancers. Cancer Cell. 34:427–438.e6. 2018. View Article : Google Scholar : PubMed/NCBI

33 

Shah SP, Roth A, Goya R, Oloumi A, Ha G, Zhao Y, Turashvili G, Ding J, Tse K, Haffari G, et al: The clonal and mutational evolution spectrum of primary triple-negative breast cancers. Nature. 486:395–399. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Tan J, Ong CK, Lim WK, Ng CC, Thike AA, Ng LM, Rajasegaran V, Myint SS, Nagarajan S, Thangaraju S, et al: Genomic landscapes of breast fibroepithelial tumors. Nat Genet. 47:1341–1345. 2015. View Article : Google Scholar : PubMed/NCBI

35 

Jézéquel P, Campone M, Gouraud W, Guérin-Charbonnel C, Leux C, Ricolleau G and Campion L: bc-GenExMiner: An easy-to-use online platform for gene prognostic analyses in breast cancer. Breast Cancer Res Treat. 131:765–775. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Jézéquel P, Frénel JS, Campion L, Guérin-Charbonnel C, Gouraud W, Ricolleau G and Campone M: bc-GenExMiner 3.0: New mining module computes breast cancer gene expression correlation analyses. Database (Oxford). 2013:bas0602013. View Article : Google Scholar : PubMed/NCBI

37 

Luo J, Wang Z, Huang J, Yao Y, Sun Q, Wang J, Shen Y, Xu L and Ren B: HOXC13 promotes proliferation of esophageal squamous cell carcinoma via repressing transcription of CASP3. Cancer Sci. 109:317–329. 2018. View Article : Google Scholar : PubMed/NCBI

38 

Marcinkiewicz KM and Gudas LJ: Altered histone mark deposition and DNA methylation at homeobox genes in human oral squamous cell carcinoma. J Cell Physiol. 229:1405–1416. 2014. View Article : Google Scholar : PubMed/NCBI

39 

Tatangelo F, Di Mauro A, Scognamiglio G, Aquino G, Lettiero A, Delrio P, Avallone A, Cantile M and Botti G: Posterior HOX genes and HOTAIR expression in the proximal and distal colon cancer pathogenesis. J Transl Med. 16:3502018. View Article : Google Scholar : PubMed/NCBI

40 

Robinson GW: Identification of signaling pathways in early mammary gland development by mouse genetics. Breast Cancer Res. 6:105–108. 2004. View Article : Google Scholar : PubMed/NCBI

41 

Tickle C, Crawley A and Goodman M: Mechanisms of invasiveness of epithelial tumours: Ultrastructure of the interactions of carcinoma cells with embryonic mesenchyme and epithelium. J Cell Sci. 33:133–155. 1978.PubMed/NCBI

42 

Morgan R and El-Tanani M: HOX genes as potential markers of circulating tumour cells. Curr Mol Med. 16:322–327. 2016. View Article : Google Scholar : PubMed/NCBI

43 

Kamkar F, Xaymardan M and Asli NS: Hox-mediated spatial and temporal coding of stem cells in homeostasis and neoplasia. Stem Cells Dev. 25:1282–1289. 2016. View Article : Google Scholar : PubMed/NCBI

44 

Nunes FD, de Almeida FC, Tucci R and de Sousa SC: Homeobox genes: A molecular link between development and cancer. Pesqui Odontol Bras. 17:94–98. 2003. View Article : Google Scholar : PubMed/NCBI

45 

Godwin AR and Capecchi MR: Hoxc13 mutant mice lack external hair. Genes Dev. 12:11–20. 1998. View Article : Google Scholar : PubMed/NCBI

46 

Comelli L, Marchetti L, Arosio D, Riva S, Abdurashidova G, Beltram F and Falaschi A: The homeotic protein HOXC13 is a member of human DNA replication complexes. Cell Cycle. 8:454–459. 2009. View Article : Google Scholar : PubMed/NCBI

47 

La Starza R, Trubia M, Crescenzi B, Matteucci C, Negrini M, Martelli MF, Pelicci PG and Mecucci C: Human homeobox gene HOXC13 is the partner of NUP98 in adult acute myeloid leukemia with t(11;12)(p15;q13). Genes Chromosomes Cancer. 36:420–423. 2003. View Article : Google Scholar : PubMed/NCBI

48 

Gurevich RM, Aplan PD and Humphries RK: NUP98-topoisomerase I acute myeloid leukemia-associated fusion gene has potent leukemogenic activities independent of an engineered catalytic site mutation. Blood. 104:1127–1136. 2004. View Article : Google Scholar : PubMed/NCBI

49 

Cantile M, Scognamiglio G, Anniciello A, Farina M, Gentilcore G, Santonastaso C, Fulciniti F, Cillo C, Franco R, Ascierto PA and Botti G: Increased HOX C13 expression in metastatic melanoma progression. J Transl Med. 10:912012. View Article : Google Scholar : PubMed/NCBI

50 

Woo CJ and Kingston RE: HOTAIR lifts noncoding RNAs to new levels. Cell. 129:1257–1259. 2007. View Article : Google Scholar : PubMed/NCBI

51 

Rinn JL, Kertesz M, Wang JK, Squazzo SL, Xu X, Brugmann SA, Goodnough LH, Helms JA, Farnham PJ, Segal E and Chang HY: Functional demarcation of active and silent chromatin domains in human HOX loci by noncoding RNAs. Cell. 129:1311–1323. 2007. View Article : Google Scholar : PubMed/NCBI

52 

Li J, Wang J, Zhong Y, Guo R, Chu D, Qiu H and Yuan Z: HOTAIR: A key regulator in gynecologic cancers. Cancer Cell Int. 17:652017. View Article : Google Scholar : PubMed/NCBI

53 

Zhang JX, Han L, Bao ZS, Wang YY, Chen LY, Yan W, Yu SZ, Pu PY, Liu N, You YP, et al: HOTAIR, a cell cycle-associated long noncoding RNA and a strong predictor of survival, is preferentially expressed in classical and mesenchymal glioma. Neuro Oncol. 15:1595–1603. 2013. View Article : Google Scholar : PubMed/NCBI

54 

Zhao W, An Y, Liang Y and Xie XW: Role of HOTAIR long noncoding RNA in metastatic progression of lung cancer. Eur Rev Med Pharmacol Sci. 18:1930–1936. 2014.PubMed/NCBI

55 

Weidle UH, Birzele F, Kollmorgen G and Ruger R: Long non-coding RNAs and their role in metastasis. Cancer Genomics Proteomics. 14:143–160. 2017. View Article : Google Scholar : PubMed/NCBI

56 

Zhou Y, Zang Y, Yang Y, Xiang J and Chen Z: Candidate genes involved in metastasis of colon cancer identified by integrated analysis. Cancer Med. 8:2338–2347. 2019. View Article : Google Scholar : PubMed/NCBI

57 

Pilato B, Pinto R, De Summa S, Lambo R, Paradiso A and Tommasi S: HOX gene methylation status analysis in patients with hereditary breast cancer. J Hum Genet. 58:51–53. 2013. View Article : Google Scholar : PubMed/NCBI

58 

Zhong Z, Shan M, Wang J, Liu T, Xia B, Niu M, Ren Y and Pang D: HOXD13 methylation status is a prognostic indicator in breast cancer. Int J Clin Exp Pathol. 8:10716–10724. 2015.PubMed/NCBI

59 

Xia B, Shan M, Wang J, Zhong Z, Geng J, He X, Vu T, Zhang D and Pang D: Homeobox A11 hypermethylation indicates unfavorable prognosis in breast cancer. Oncotarget. 8:9794–9805. 2017.PubMed/NCBI

60 

Milevskiy MJ, Al-Ejeh F, Saunus JM, Northwood KS, Bailey PJ, Betts JA, McCart Reed AE, Nephew KP, Stone A, Gee JM, et al: Long-range regulators of the lncRNA HOTAIR enhance its prognostic potential in breast cancer. Hum Mol Genet. 25:3269–3283. 2016. View Article : Google Scholar : PubMed/NCBI

61 

Botti G, De Chiara A, Di Bonito M, Cerrone M, Malzone MG, Collina F and Cantile M: Noncoding RNAs within the HOX gene network in tumor pathogenesis and progression. J Cell Physiol. 234:395–413. 2018. View Article : Google Scholar : PubMed/NCBI

62 

Sun Z, Wu XY and Wu CL: The association between LncRNA HOTAIR and cancer lymph node metastasis and distant metastasis: A meta-analysis. Neoplasma. 65:178–184. 2018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2020
Volume 19 Issue 1

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li C, Cui J, Zou L, Zhu L and Wei W: Bioinformatics analysis of the expression of HOXC13 and its role in the prognosis of breast cancer . Oncol Lett 19: 899-907, 2020
APA
Li, C., Cui, J., Zou, L., Zhu, L., & Wei, W. (2020). Bioinformatics analysis of the expression of HOXC13 and its role in the prognosis of breast cancer . Oncology Letters, 19, 899-907. https://doi.org/10.3892/ol.2019.11140
MLA
Li, C., Cui, J., Zou, L., Zhu, L., Wei, W."Bioinformatics analysis of the expression of HOXC13 and its role in the prognosis of breast cancer ". Oncology Letters 19.1 (2020): 899-907.
Chicago
Li, C., Cui, J., Zou, L., Zhu, L., Wei, W."Bioinformatics analysis of the expression of HOXC13 and its role in the prognosis of breast cancer ". Oncology Letters 19, no. 1 (2020): 899-907. https://doi.org/10.3892/ol.2019.11140