Research article

Downregulation of CDC14B in 5218 breast cancer patients: A novel prognosticator for triple-negative breast cancer

  • Received: 01 September 2020 Accepted: 02 November 2020 Published: 13 November 2020
  • Breast cancer is the most common female malignancy worldwide and the prognosis of triple-negative breast cancer (TNBC) and advanced breast cancer patients is unsatisfying. The exploration of novel prognostic indicators and appropriate targets is crucial for improving the treatment outcomes of breast cancer patients. The cell division cycle protein 14B (CDC14B) is known for its roles in cell cycle control, but its expression status and molecular function in breast cancer is unknown. This study explores the expression patterns and clinical values of CDC14B in breast cancer tissues. For this research, the authors downloaded gene microarrays and RNA sequencing datasets to examine the expression levels of CDC14B in 5218 breast cancer tissues, comparing them to the expression levels in 1176 normal breast tissues. The relationships between CDC14B and clinicopathologic characteristics of breast cancer were also addressed. The mutation conditions of CDC14B were then clarified using cBioPortal. Finally, differentially expressed genes and co-expressed genes related to CDC14B were filtered using the Limma-Voom package. These genes were intersected to conduct functional annotations and to construct a protein-protein interaction network. It was observed that CDC14B was significantly downregulated in breast cancer tissues but not in normal breast tissues (standardized mean difference = -1.17 [-1.50--0.85], area under the curve = 0.88). In addition, CDC14B downregulation was correlated with the poor prognosis of TNBC patients (hazard ratios < 1; p < 0.05). Amplification was detected to be the most frequent alteration of CDC14B. The presence of this alteration forecasted unfavourable overall survival outcomes in breast cancer patients (p < 0.05). Dysregulated genes that co-expressed with CDC14B were pivotal in cell cycle (namely mitotic-nuclear division and DNA packaging complex) and cancer-related signaling pathways (namely the peroxisome proliferators activated receptor [PPAR] signalling pathway and the AMP-activated protein kinase [AMPK] signalling pathway). Moreover, the genes ADIPOQ and CCNE2 were identified as two promising prognostic factors in breast cancer. In summary, CDC14B was downregulated in breast cancer tissue and may be a promising hallmark in TNBC patients. The dysregulated genes co-expressed with CDC14B may play an important role in the development of breast cancer through PPAR and AMPK signalling pathways.

    Citation: Jian-Di Li, Gang Chen, Mei Wu, Yu Huang, Wei Tang. Downregulation of CDC14B in 5218 breast cancer patients: A novel prognosticator for triple-negative breast cancer[J]. Mathematical Biosciences and Engineering, 2020, 17(6): 8152-8181. doi: 10.3934/mbe.2020414

    Related Papers:

  • Breast cancer is the most common female malignancy worldwide and the prognosis of triple-negative breast cancer (TNBC) and advanced breast cancer patients is unsatisfying. The exploration of novel prognostic indicators and appropriate targets is crucial for improving the treatment outcomes of breast cancer patients. The cell division cycle protein 14B (CDC14B) is known for its roles in cell cycle control, but its expression status and molecular function in breast cancer is unknown. This study explores the expression patterns and clinical values of CDC14B in breast cancer tissues. For this research, the authors downloaded gene microarrays and RNA sequencing datasets to examine the expression levels of CDC14B in 5218 breast cancer tissues, comparing them to the expression levels in 1176 normal breast tissues. The relationships between CDC14B and clinicopathologic characteristics of breast cancer were also addressed. The mutation conditions of CDC14B were then clarified using cBioPortal. Finally, differentially expressed genes and co-expressed genes related to CDC14B were filtered using the Limma-Voom package. These genes were intersected to conduct functional annotations and to construct a protein-protein interaction network. It was observed that CDC14B was significantly downregulated in breast cancer tissues but not in normal breast tissues (standardized mean difference = -1.17 [-1.50--0.85], area under the curve = 0.88). In addition, CDC14B downregulation was correlated with the poor prognosis of TNBC patients (hazard ratios < 1; p < 0.05). Amplification was detected to be the most frequent alteration of CDC14B. The presence of this alteration forecasted unfavourable overall survival outcomes in breast cancer patients (p < 0.05). Dysregulated genes that co-expressed with CDC14B were pivotal in cell cycle (namely mitotic-nuclear division and DNA packaging complex) and cancer-related signaling pathways (namely the peroxisome proliferators activated receptor [PPAR] signalling pathway and the AMP-activated protein kinase [AMPK] signalling pathway). Moreover, the genes ADIPOQ and CCNE2 were identified as two promising prognostic factors in breast cancer. In summary, CDC14B was downregulated in breast cancer tissue and may be a promising hallmark in TNBC patients. The dysregulated genes co-expressed with CDC14B may play an important role in the development of breast cancer through PPAR and AMPK signalling pathways.


    加载中


    [1] F. Bray, J. Ferlay, I. Soerjomataram, R. L. Siegel, L. A. Torre, A. Jemal, Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries, CA Cancer J. Clin., 68 (2018), 394-424. doi: 10.3322/caac.21492
    [2] R. L. Siegel, K. D. Miller, A. Jemal, Cancer statistics, CA Cancer J. Clin., 70 (2020), 7-30. doi: 10.3322/caac.21590
    [3] C. Larsson, A. Ehinger, S. Winslow, K. Leandersson, M. Klintman, L. Dahl, et al., Prognostic implications of the expression levels of different immunoglobulin heavy chain-encoding RNAs in early breast cancer, NPJ Breast Cancer, 6 (2020), 28. doi: 10.1038/s41523-020-0170-2
    [4] M. Alsaleem, M. S. Toss, C. Joseph, M. Aleskandarany, S. Kurozumi, I. Alshankyty, et al., The molecular mechanisms underlying reduced E-cadherin expression in invasive ductal carcinoma of the breast: high throughput analysis of large cohorts, Mod. Pathol., 32 (2019), 967-976. doi: 10.1038/s41379-019-0209-9
    [5] S. Yoon, H. S. Won, K. Kang, K. Qiu, W. J. Park, Y. H. Ko, Hormone Receptor-Status Prediction in Breast Cancer Using Gene Expression Profiles and Their Macroscopic Landscape, Cancers, 12 (2020), 1165. doi: 10.3390/cancers12051165
    [6] A. Nasir, M. M. H. Bullo, Z. Ahmed, A. Imtiaz, E. Yaqoob, M. Jadoon, et al., Nutrigenomics: Epigenetics and cancer prevention: A comprehensive review, Crit. Rev. Food Sci. Nutr., 60 (2020), 1375-1387. doi: 10.1080/10408398.2019.1571480
    [7] D. R. Chen, W. C. Hsieh, Y. L. Liao, K. J. Lin, Y. F. Wang, P. H. Lin, Imbalances in the disposition of estrogen and naphthalene in breast cancer patients: a potential biomarker of breast cancer risk, Sci. Rep., 10 (2020), 11773. doi: 10.1038/s41598-020-68814-5
    [8] D. Ma, Y. Z. Jiang, Y. Xiao, M. D. Xie, S. Zhao, X. Jin, et al., Integrated molecular profiling of young and elderly patients with triple-negative breast cancer indicates different biological bases and clinical management strategies, Cancer, 126 (2020), 3209-3218. doi: 10.1002/cncr.32922
    [9] J. Chen, M. K. Haanpaa, J. J. Gruber, N. Jager, J. M. Ford, M. P. Snyder, High-Resolution Bisulfite-Sequencing of Peripheral Blood DNA Methylation in Early-Onset and Familial Risk Breast Cancer Patients, Clin. Cancer Res., 25 (2019), 5301-5314. doi: 10.1158/1078-0432.CCR-18-2423
    [10] I. V. Ulasov, A. V. Borovjagin, P. Timashev, M. Cristofanili, D. R. Welch, KISS1 in breast cancer progression and autophagy, Cancer Metastasis Rev., 38 (2019), 493-506. doi: 10.1007/s10555-019-09814-4
    [11] M. S. M. Issac, E. Yousef, M. R. Tahir, L. A. Gaboury, MCM2, MCM4, and MCM6 in Breast Cancer: Clinical Utility in Diagnosis and Prognosis, Neoplasia (N. Y., NY, U. S.), 21 (2019), 1015-1035. doi: 10.1016/j.neo.2019.07.011
    [12] Q. Wu, B. Li, Z. Li, J. Li, S. Sun, S. Sun, Cancer-associated adipocytes: key players in breast cancer progression, J. Hematol. Oncol., 12 (2019), 95. doi: 10.1186/s13045-019-0778-6
    [13] R. Eyre, D. G. Alferez, A. Santiago-Gomez, K. Spence, J. C. McConnell, C. Hart, et al., Microenvironmental IL1β promotes breast cancer metastatic colonisation in the bone via activation of Wnt signalling, Nat. Commun., 10 (2019), 5016. doi: 10.1038/s41467-019-12807-0
    [14] K. Pandey, H. J. An, S. K. Kim, S. A. Lee, S. Kim, S. M. Lim, et al., Molecular mechanisms of resistance to CDK4/6 inhibitors in breast cancer: A review, Int. J. Cancer, 145 (2019), 1179-1188. doi: 10.1002/ijc.32020
    [15] K. Goutsouliak, J. Veeraraghavan, V. Sethunath, C. De Angelis, C. K. Osborne, M. F. Rimawi, et al., Towards personalized treatment for early stage HER2-positive breast cancer, Nat. Rev. Clin. Oncol., 17 (2020), 233-250. doi: 10.1038/s41571-019-0299-9
    [16] Collaborative Group on Hormonal Factors in Breast Cancer, Type and timing of menopausal hormone therapy and breast cancer risk: individual participant meta-analysis of the worldwide epidemiological evidence, Lancet, 394 (2019), 1159-1168.
    [17] G. E. Naoum, L. Salama, A. Ho, N. K. Horick, O. Oladeru, M. Abouegylah, et al., The Impact of Chest Wall Boost on Reconstruction Complications and Local Control in Patients Treated for Breast Cancer, Int. J. Radiat. Oncol., Biol., Phys., 105 (2019), 155-164. doi: 10.1016/j.ijrobp.2019.04.027
    [18] A. Darlix, G. Louvel, J. Fraisse, W. Jacot, E. Brain, M. Debled, et al., Impact of breast cancer molecular subtypes on the incidence, kinetics and prognosis of central nervous system metastases in a large multicentre real-life cohort, Br. J. Cancer, 121 (2019), 1-10.
    [19] S. M. Tolaney, A. M. Wardley, S. Zambelli, J. F. Hilton, T. A. Troso-Sandoval, F. Ricci, et al., Abemaciclib plus trastuzumab with or without fulvestrant versus trastuzumab plus standard-of-care chemotherapy in women with hormone receptor-positive, HER2-positive advanced breast cancer (monarcHER): a randomised, open-label, phase 2 trial, Lancet Oncol., 21 (2020), 763-775. doi: 10.1016/S1470-2045(20)30112-1
    [20] J. Li, K. Yu, D. Pang, C. Wang, J. Jiang, S. Yang, et al., Adjuvant Capecitabine With Docetaxel and Cyclophosphamide Plus Epirubicin for Triple-Negative Breast Cancer (CBCSG010): An Open-Label, Randomized, Multicenter, Phase III Trial, J. Clin. Oncol., 38 (2020), 1774-1784.
    [21] P. Sun, D. Zhang, H. Huang, Y. Yu, Z. Yang, Y. Niu, et al., MicroRNA-1225-5p acts as a tumor-suppressor in laryngeal cancer via targeting CDC14B, Biol. Chem., 400 (2019), 237-246. doi: 10.1515/hsz-2018-0265
    [22] S. Ovejero, P. Ayala, M. Malumbres, F. X. Pimentel-Muinos, A. Bueno, M. P. Sacristan, Biochemical analyses reveal amino acid residues critical for cell cycle-dependent phosphorylation of human Cdc14A phosphatase by cyclin-dependent kinase 1, Sci. Rep., 8 (2018), 11871. doi: 10.1038/s41598-018-30253-8
    [23] M. Dietachmayr, A. Rathakrishnan, O. Karpiuk, F. von Zweydorf, T. Engleitner, V. Fernandez-Saiz, et al., Antagonistic activities of CDC14B and CDK1 on USP9X regulate WT1-dependent mitotic transcription and survival, Nat. Commun., 11 (2020), 1268. doi: 10.1038/s41467-020-15059-5
    [24] H. Lin, K. Ha, G. Lu, X. Fang, R. Cheng, Q. Zuo, et al., Cdc14A and Cdc14B Redundantly Regulate DNA Double-Strand Break Repair, Mol. Cell. Biol., 35 (2015), 3657-3668. doi: 10.1128/MCB.00233-15
    [25] Y. Wang, J. Tian, C. Huang, J. Ma, G. Hu, Y. Chen, et al., P53 suppresses SENP3 phosphorylation to mediate G2 checkpoint, Cell Discovery, 6 (2020), 21.
    [26] Y. Kim, J. W. Choi, J. H. Lee, Y. S. Kim, Loss of CDC14B expression in clear cell renal cell carcinoma: meta-analysis of microarray data sets, Am. J. Clin. Pathol., 141 (2014), 551-558. doi: 10.1309/AJCP4PE4JPSRGBQS
    [27] D. M. Kokkinakis, X. Liu, S. Chada, M. M. Ahmed, M. M. Shareef, U. K. Singha, et al., Modulation of gene expression in human central nervous system tumors under methionine deprivation-induced stress, Cancer Res., 64 (2004), 7513-7525. doi: 10.1158/0008-5472.CAN-04-0592
    [28] D. F. Calvisi, S. Ladu, F. Pinna, M. Frau, M. L. Tomasi, M. Sini, et al., SKP2 and CKS1 promote degradation of cell cycle regulators and are associated with hepatocellular carcinoma prognosis, Gastroenterology, 137 (2009), 1816-1826. doi: 10.1053/j.gastro.2009.08.005
    [29] B. Wu, X. Chen, J. Wang, X. Qing, Z. Wang, X. Ding, et al., Separation and characterization of extracellular vesicles from human plasma by asymmetrical flow field-flow fractionation, Anal. Chim. Acta, 1127 (2020), 234-245. doi: 10.1016/j.aca.2020.06.071
    [30] L. Tu, R. Guan, H. Yang, Y. Zhou, W. Hong, L. Ma, et al., Assessment of the expression of the immune checkpoint molecules PD-1, CTLA4, TIM-3 and LAG-3 across different cancers in relation to treatment response, tumor-infiltrating immune cells and survival, Int. J. Cancer, 147 (2020), 423-439. doi: 10.1002/ijc.32785
    [31] M. J. Goldman, J. Zhang, N. A. Fonseca, I. Cortes-Ciriano, Q. Xiang, B. Craft, et al., A user guide for the online exploration and visualization of PCAWG data, Nat. Commun., 11 (2020), 3400. doi: 10.1038/s41467-020-16785-6
    [32] E. Vagia, D. Mahalingam, M. Cristofanilli, The Landscape of Targeted Therapies in TNBC, Cancers, 12 (2020), 916. doi: 10.3390/cancers12040916
    [33] R. Yang, L. Xing, M. Wang, H. Chi, L. Zhang, J. Chen, Comprehensive Analysis of Differentially Expressed Profiles of lncRNAs/mRNAs and miRNAs with Associated ceRNA Networks in Triple-Negative Breast Cancer, Cell. Physiol. Biochem., 50 (2018), 473-488. doi: 10.1159/000494162
    [34] J. Wang, Y. Su, Y. Tian, Y. Ding, X. Wang, Characterization of DNA hydroxymethylation profile in cervical cancer, Artif. Cells, Nanomed., Biotechnol., 47 (2019), 2706-2714. doi: 10.1080/21691401.2019.1634578
    [35] M. N. H. Luijten, J. X. T. Lee, K. C. Crasta, Mutational game changer: Chromothripsis and its emerging relevance to cancer, Mutat. Res., 777 (2018), 29-51. doi: 10.1016/j.mrrev.2018.06.004
    [36] D. A. Quigley, H. X. Dang, S. G. Zhao, P. Lloyd, R. Aggarwal, J. J. Alumkal, et al., Genomic Hallmarks and Structural Variation in Metastatic Prostate Cancer, Cell, 174 (2018), 758-769. doi: 10.1016/j.cell.2018.06.039
    [37] A. Marcozzi, F. Pellestor, W. P. Kloosterman, The Genomic Characteristics and Origin of Chromothripsis, Chromothripsis, Humana Press, New York, NY, 1769 (2018), 3-9.
    [38] The ICGC/TCGA Pan-Cancer Analysis of Whole Genomes Consortium, Pan-cancer analysis of whole genomes, Nature, 578 (2020), 82-93.
    [39] N. Voronina, J. K. L. Wong, D. Hubschmann, M. Hlevnjak, S. Uhrig, C. E. Heilig, et al., The landscape of chromothripsis across adult cancer types, Nat. Commun., 11 (2020), 2320. doi: 10.1038/s41467-020-16134-7
    [40] Z. Li, X. Zhang, C. Hou, Y. Zhou, J. Chen, H. Cai, et al., Comprehensive identification and characterization of somatic copy number alterations in triple‑negative breast cancer, Int. J. Oncol., 56 (2020), 522-530.
    [41] F. Hong, S. Pan, Y. Guo, P. Xu, Y. Zhai, PPARs as nuclear receptors for nutrient and energy metabolism, Molecules, 24 (2019), 2545. doi: 10.3390/molecules24142545
    [42] W. H. Chang, A. G. Lai, An integrative pan-cancer investigation reveals common genetic and transcriptional alterations of AMPK pathway genes as important predictors of clinical outcomes across major cancer types, BMC cancer, 20 (2020), 773. doi: 10.1186/s12885-020-07286-2
    [43] Y. Liu, J. K. Colby, X. Zuo, J. Jaoude, D. Wei, I. Shureiqi, The role of PPAR-δ in metabolism, inflammation, and cancer: Many characters of a critical transcription factor, Int. J. Mol. Sci., 19 (2018), 3339. doi: 10.3390/ijms19113339
    [44] W. H. Chang, A. G. Lai, The pan-cancer mutational landscape of the PPAR pathway reveals universal patterns of dysregulated metabolism and interactions with tumor immunity and hypoxia, Ann. N. Y. Acad. Sci., 1448 (2019), 65-82. doi: 10.1111/nyas.14170
    [45] G. Sultan, S. Zubair, I. A. Tayubi, H. U. Dahms, I. H. Madar, Towards the early detection of ductal carcinoma (a common type of breast cancer) using biomarkers linked to the PPAR(γ) signaling pathway, Bioinformation, 15 (2019), 799-805. doi: 10.6026/97320630015799
    [46] Y. P. Hwang, S. S. Won, S. W. Jin, G. H. Lee, T. H. Pham, J. H. Choi, et al., WY-14643 regulates CYP1B1 expression through peroxisome proliferator-activated receptor α-mediated signaling in human breast cancer cells, Int. J. Mol. Sci., 20 (2019), 5928. doi: 10.3390/ijms20235928
    [47] L. Ponnusamy, S. R. Natarajan, K. Thangaraj, R. Manoharan, Therapeutic aspects of AMPK in breast cancer: Progress, challenges, and future directions, Biochim. Biophys. Acta, Rev. Cancer, 1874 (2020), 188379. doi: 10.1016/j.bbcan.2020.188379
    [48] S. C. Kwong, A. H. A. Jamil, A. Rhodes, N. A. Taib, I. Chung, Metabolic role of fatty acid binding protein 7 in mediating triple-negative breast cancer cell death via PPAR-α signaling, J. Lipid Res., 60 (2019), 1807-1817. doi: 10.1194/jlr.M092379
    [49] M. K. Apaya, P. W. Hsiao, Y. C. Yang, L. F. Shyur, Deregulating the CYP2C19/epoxy-eicosatrienoic acid-associated FABP4/FABP5 signaling network as a therapeutic approach for metastatic triple-negative breast cancer, Cancers, 12 (2020), 199. doi: 10.3390/cancers12010199
    [50] H. Liu, H. Sun, B. Zhang, S. Liu, S. Deng, Z. Weng, et al., F-FDG PET imaging for monitoring the early anti-tumor effect of albendazole on triple-negative breast cancer, Breast cancer, 27 (2020), 372-380. doi: 10.1007/s12282-019-01027-5
    [51] H. Fang, G. Du, Q. Wu, R. Liu, C. Chen, J. Feng, HDAC inhibitors induce proline dehydrogenase (POX) transcription and anti-apoptotic autophagy in triple negative breast cancer, Acta Biochim. Biophys. Sin., 51 (2019), 1064-1070. doi: 10.1093/abbs/gmz097
    [52] D. Zhou, X. Zhao, M. Yu, Y. Xu, C. Fu, K. Zheng, et al., Anti-migration and anti-invasion effects of 2-hydroxy-6-tridecylbenzoic acid is associated with the enhancement of CYP1B1 expression through activating the AMPK signaling pathway in triple-negative breast cancer cells, Nat. Prod. Res., 2020 (2020), 1-5.
    [53] N. Bhuiyan, P. Singh, S. M. Harden, S. K. Mama, Rural physical activity interventions in the United States: a systematic review and RE-AIM evaluation, Int. J. Behav. Nutr. Phys. Act., 16 (2019), 140. doi: 10.1186/s12966-019-0903-5
    [54] G. Pasello, A. Pavan, I. Attili, A. Bortolami, L. Bonanno, J. Menis, et al., Real world data in the era of Immune Checkpoint Inhibitors (ICIs): Increasing evidence and future applications in lung cancer, Cancer Treat Rev., 87 (2020), 102031. doi: 10.1016/j.ctrv.2020.102031
    [55] I. Meisingset, O. Vasseljen, N. K. Vøllestad, H. S. Robinson, A. Woodhouse, K. B. Engebretsen, et al., Novel approach towards musculoskeletal phenotypes, Eur. J. Pain, 24 (2020), 921-932. doi: 10.1002/ejp.1541
    [56] C. Lee, K. J. Fernandez, S. Alexandrou, C. M. Sergio, N. Deng, S. Rogers, et al., Cyclin E2 Promotes Whole Genome Doubling in Breast Cancer, Cancers, 12 (2020), 2268. doi: 10.3390/cancers12082268
  • MBE-17-06-414-Supplementary.pdf
  • Reader Comments
  • © 2020 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(3899) PDF downloads(152) Cited by(3)

Article outline

Figures and Tables

Figures(13)  /  Tables(1)

Other Articles By Authors

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog