Evidence for Tumour Suppressor Function of DOK7 in Human Breast Cancer

Abstract

Introduction: Downstream of tyrosine kinase 7 (DOK-7) is a member of the DOK family, which has been associated with the development and progression of various humancancers. Previously, identification of CpG hypermethylation in DOK-7 promoter was identified in breast cancer. Method: DOK-7 mRNA extraction and reverse transcription were performed on fresh frozen breast cancer tissue samples and normal background breast tissue. Transcript levels of expression were analyzed against TNM stage, tumour grade and clinical outcome over a 10-year follow-up period. Results: Levels of DOK-7 expression decreased significantly with increasing TNM stage. Higher DOK-7 expression was correlated with longer disease free and overall survival times. Conclusion: To our knowledge, this is the first study to investigate DOK-7 expression in human breast cancer. We identify a potential DOK-7 tumour suppressor role. DOK-7 as a prognostic biomarker in human breast cancer should be included in future validation studies.

Share and Cite:

J. Bracken, T. Ghanem, A. Kasem, W. Jiang and K. Mokbel, "Evidence for Tumour Suppressor Function of DOK7 in Human Breast Cancer," Journal of Cancer Therapy, Vol. 5 No. 1, 2014, pp. 67-73. doi: 10.4236/jct.2014.51009.

Conflicts of Interest

The authors declare no conflicts of interest.

References

[1] S. Mehta, et al., “Predictive and Prognostic Molecular Markers for Cancer Medicine,” Therapeutic Advances in Medical Oncology, Vol. 2, No. 2, 2010, pp. 125-148. http://dx.doi.org/10.1177/175 8834009360519
[2] A. W. Opstal-van Winden, et al., “Searching for Early Breast Cancer Biomarkers by Serum Protein Profiling of Pre-Diagnostic Serum; a Nested Case-Control Study,” BMC Cancer, Vol. 11, 2011, p. 381. http://dx.doi.org/10.1186/1471-2407-11-381
[3] S. Y. Park, et al., “Promoter CpG Island Hypermethylation during Breast Cancer Progression,” Virchows Arch, Vol. 458, No. 1, 2011, pp. 73-84. http://dx.doi.org/10.1007/s00428-010-1013-6
[4] P. Taneja, et al., “Classical and Novel Prognostic Markers for Breast Cancer and their Clinical Significance,” Clinical Medicine Insights: Oncology, Vol. 4, 2010, pp. 15-34.
[5] M. T. Weigel and M. Dowsett, “Current and Emerging Biomarkers in Breast Cancer: Prognosis and Prediction,” Endocrine-Related Cancer, Vol. 17, No. 4, 2010, pp. R245-R262. http://dx.doi.org/10. 1677/ERC-10-0136
[6] F. Zhang, et al., “Recursive SVM Biomarker Selection for Early Detection of Breast Cancer in Peripheral Blood,” BMC Medical Genomics, Vol. 6, Suppl. 1, 2013, p. S4. http://dx.doi.org/10.1186/ 1755-8794-6-S1-S4
[7] K. D. Cole, H. J. He and L. Wang, “Breast Cancer Biomarker Measurements and Standards,” PROTEOMICSClinical Applications, Vol. 7, No. 1-2, 2013, pp. 17-29. http://dx.doi.org/10.1002/prca. 201200075
[8] A. Bedirian, et al., “Pleckstrin Homology and Phosphotyrosine-Binding Domain-Dependent Membrane Association and Tyrosine Phosphorylation of Dok-4, an Inhibitory Adapter Molecule Expressed in Epithelial Cells,” Journal of Biological Chemistry, Vol. 279, 2004, pp. 19335-19349. http://dx.doi.org/ 10.1074/jbc.M310689200
[9] A. H. Berger, et al., “Identification of DOK Genes as Lung Tumor Suppressors,” Nature Genetics, Vol. 42, 2010, pp. 216-223. http://dx.doi.org/10.1038/ng.527
[10] C. H. An, et al., “Mutational and Expressional Analysis of a Haploinsufficient Tumor Suppressor Gene DOK2 in Gastric and Colorectal Cancers,” APMIS, Vol. 119, No. 8, 2011, pp. 562-564.
http://dx.doi.org/10.1111/j.1600-0463.2011.02749.x
[11] H. Miyagaki, et al., “DOK2 as a Marker of Poor Prognosis of Patients with Gastric Adenocarcinoma after Curative Resection,” Annals of Surgical Oncology, Vol. 19, No. 5, 2012, pp. 1560-1567.
http://dx.doi.org/10.1245/s10434-011-2157-6
[12] R. Mashima, et al., “Mice Lacking Dok-1, Dok-2, and Dok-3 Succumb to Aggressive Histiocytic Sarcoma,” Laboratory Investigation, Vol. 90, 2010, pp. 1357-1364. http://dx.doi.org/10.1038/ labinvest.2010.121
[13] R. J. Crowder, et al., “Dok-6, a Novel p62 Dok Family Member, Promotes Ret-Mediated Neurite Outgrowth,” Journal of Biological Chemistry, Vol. 279, 2004, pp. 42072-42081. http://dx.doi.org/10. 1074/jbc.M403726200
[14] J. Grimm, et al., “Novel p62dok Family Members, dok-4 and dok-5, Are Substrates of the c-Ret Receptor Tyrosine Kinase and Mediate Neuronal Differentiation,” Journal of Cell Biology, Vol. 154, No. 2, 2001, pp. 345-354. http://dx.doi.org/10.1083/jcb.200102032
[15] J. Cossins, et al., “The Spectrum of Mutations That Underlie the Neuromuscular Junction Synaptopathy in DOK7 Congenital Myasthenic Syndrome,” Human Molecular Genetics, Vol. 21, No. 17, 2012, pp. 3765-3775. http://dx.doi.org/10.1093/hmg/dds198
[16] E. Bergamin, et al., “The Cytoplasmic Adaptor Protein Dok7 Activates the Receptor Tyrosine Kinase MuSK via Dimerization,” Molecular Cell, Vol. 39, No. 1, 2010, pp. 100-109. http://dx.doi.org/10.1016/ j.molcel.2010.06.007
[17] A. Inoue, et al., “Dok-7 Activates the Muscle Receptor Kinase MuSK and Shapes Synapse Formation,” Science Signaling, Vol. 2, No. 59, 2009, p. ra7. http://dx.doi.org/10.1126/scisignal.2000113
[18] A. Klein, et al., “DOK7 Congenital Myasthenic Syndrome in Childhood: Early Diagnostic Clues in 23 Children,” Neuromuscular Disorders, Vol. 23, No. 11, 2013, pp. 883-891. http://dx.doi.org/10.1016/ j.nmd.2013.06.002
[19] J. S. Muller, et al., “Phenotypical Spectrum of DOK7 Mutations in Congenital Myasthenic Syndromes,” Brain, Vol. 130, No. 6, 2007, pp. 1497-1506. http://dx.doi.org/10.1093/brain/awm068
[20] K. Okada, et al., “The Muscle Protein Dok-7 Is Essential for Neuromuscular Synaptogenesis,” Science, Vol. 312, No. 5781, 2006, pp. 1802-1805. http://dx.doi.org/10.1126/science.1127142
[21] T. Cheusova, et al., “Casein Kinase 2-Dependent Serine Phosphorylation of MuSK Regulates Acetylcholine Receptor Aggregation at the Neuromuscular Junction,” Genes & Development, Vol. 20, 2006, pp. 1800-1816. http://dx.doi.org/10.1101/gad.375206
[22] D. J. Glass, et al., “Agrin Acts via a MuSK Receptor Complex,” Cell, Vol. 85, No. 4, 1996, pp. 513-523. http://dx.doi.org/10.1016/S0092-8674(00)81252-0
[23] S. Klein-Scory, et al., “Immunoscreening of the Extracellular Proteome of Colorectal Cancer Cells,” BMC Cancer, Vol. 10, 2010, p. 70. http://dx.doi.org/10.1186/1471-2407-10-70
[24] D. Drygin, et al., “Protein Kinase CK2 Modulates IL-6 Expression in Inflammatory Breast Cancer,” Biochemical and Biophysical Research Communications, Vol. 415, No. 1, 2011, pp. 163-167. http://dx.doi.org/10.1016/j.bbrc.2011.10.046
[25] H. Heyn, et al., “DNA Methylation Profiling in Breast Cancer Discordant Identical Twins Identifies DOK7 as Novel Epigenetic Biomarker,” Carcinogenesis, Vol. 34, No. 1, 2013, pp. 102-108. http://dx.doi.org/10.1093/carcin/bgs321
[26] M. Esteller, “Epigenetics in Cancer,” New England Journal of Medicine, Vol. 358, 2008, pp. 1148-1159. http://dx.doi.org/10.1056/NEJMra072067
[27] Y. Huang, et al., “Epigenetics in Breast Cancer: What’s New?” Breast Cancer Research, Vol. 13, 2011, p. 225. http://dx.doi.org/10.1186/bcr2925
[28] S. Sharma, T. K. Kelly and P. A. Jones, “Epigenetics in Cancer,” Carcinogenesis, Vol. 31, No. 1, 2010, pp. 27-36. http://dx.doi.org/10.1093/carcin/bgp220
[29] J. F. Costello, et al., “Aberrant CpG-Island Methylation Has Non-Random and Tumour-Type-Specific Patterns,” Nature Genetics, Vol. 24, 2000, pp. 132-138. http://dx.doi.org/10.1038/72785
[30] J. Hamuro, et al., “The Transcription Factor Sp1 Plays a Crucial Role in dok-7 Gene Expression,” Biochemical and Biophysical Research Communications, Vol. 408, No. 2, 2011, pp. 293-299.
http://dx.doi.org/10.1016/j.bbrc.2011.04.020
[31] S. Lu and M. C. Archer, “Sp1 Coordinately Regulates de Novo Lipogenesis and Proliferation in Cancer Cells,” International Journal of Cancer, Vol. 126, No. 2, 2010, pp. 416-425. http://dx.doi.org/10.1002/ ijc.24761
[32] S. U. Mertens-Talcott, et al., “Betulinic Acid Decreases ER-Negative Breast Cancer Cell Growth in Vitro and in Vivo: Role of Sp Transcription Factors and microRNA-27a:ZBTB10,” Molecular Carcinogenesis, Vol. 52, No. 8, 2013, pp. 591-602. http://dx.doi.org/10.1002/mc.21893
[33] X. B. Wang, et al., “Expression and Prognostic Value of Transcriptional Factor sp1 in Breast Cancer,” Chinese Journal of Cancer, Vol. 26, No. 9, 2007, pp. 996-1000.
[34] M. Wei, et al., “Stat6 Cooperates with Sp1 in Controlling Breast Cancer Cell Proliferation by Modulating the Expression of p21 (Cip1/WAF1) and p27 (Kip1),” Cellular Oncology, Vol. 36, No. 1, 2013, pp. 79-93. http://dx.doi.org/10.1007/s13402-012-0115-3
[35] X. Yang, et al., “HSF1 and Sp1 Are Involved in the Regulation of FUT4 Gene Expression and Cell Proliferation in Breast Cancer Cells,” Journal of Cellular Biochemistry, Vol. 115, No. 1, 2014, pp. 168-178.
[36] L. Yue, et al., “The Oncoprotein HBXIP Activates Transcriptional Coregulatory Protein LMO4 via Sp1 to Promote Proliferation of Breast Cancer Cells,” Carcinogenesis, Vol. 34, No. 4, 2013, pp. 927-935. http://dx.doi.org/10.1093/carcin/bgs399
[37] Y. Zhang, et al., “The Oncoprotein HBXIP Upregulates PDGFB via Activating Transcription Factor Sp1 to Promote the Proliferation of Breast Cancer Cells,” Biochemical and Biophysical Research Communications, Vol. 434, No. 2, 2013, pp. 305-310. http://dx.doi.org/10.1016/j.bbrc.2013.02.123
[38] W. Sasi, et al., “Higher Expression Levels of SOCS 1,3,4,7 Are Associated with Earlier Tumour Stage and Better Clinical Outcome in Human Breast Cancer,” BMC Cancer, Vol. 10, 2010, p. 178. http://dx.doi.org/10.1186/1471-2407-10-178
[39] W. Al Sarakbi, et al., “The mRNA Expression of SETD2 in Human Breast Cancer: Correlation with ClinicoPathological Parameters,” BMC Cancer, Vol. 9, 2009, p. 290. http://dx.doi.org/10.1186/1471-2407-9-290
[40] W. G. Jiang, et al., “Prognostic Value of rho GTPases and rho Guanine Nucleotide Dissociation Inhibitors in Human Breast Cancers,” Clinical Cancer Research, Vol. 9, No. 17, 2003, pp. 6432-6440.
[41] T. Nakagawa, et al., “Decreased Expression of SOCS-3 mRNA in Breast Cancer with Lymph Node Metastasis,” Oncology Reports, Vol. 19, No. 1, 2008, pp. 33-39.
[42] Z. Dogusan, et al., “Expression of SOCS Genes in Normal and Leukemic Human Leukocytes Stimulated by Prolactin, Growth Hormone and Cytokines,” Journal of Neuroimmunology, Vol. 109, No. 1, 2000, pp. 34-39. http://dx.doi.org/10.1016/S0165-5728(00)00300-3
[43] A. Saulnier, et al., “Inactivation of the Putative Suppressor Gene DOK1 by Promoter Hypermethylation in Primary Human Cancers,” International Journal of Cancer, Vol. 130, No. 11, 2012, pp. 2484-2494. http://dx.doi.org/10.1002/ijc.26299
[44] A. Ben Ammar, et al., “A Mutation Causes MuSK Reduced Sensitivity to Agrin and Congenital Myasthenia,” PLoS One, Vol. 8, 2013, Article ID: e53826. http://dx.doi.org/10.1371/annotation/ 3ff2b918-c83c-4c6f-a2e2-4d91294ec92f
[45] W. C. Chang and J. J. Hung, “Functional Role of Post-Translational Modifications of Sp1 in Tumorigenesis,” Journal of Biomedical Science, Vol. 19, 2012, p. 94. http://dx.doi.org/10.1186/1423-0127-19-94

Copyright © 2024 by authors and Scientific Research Publishing Inc.

Creative Commons License

This work and the related PDF file are licensed under a Creative Commons Attribution 4.0 International License.