DOI QR코드

DOI QR Code

PKCδ-dependent Activation of the Ubiquitin Proteasome System is Responsible for High Glucose-induced Human Breast Cancer MCF-7 Cell Proliferation, Migration and Invasion

  • Zhu, Shan (Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University) ;
  • Yao, Feng (Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University) ;
  • Li, Wen-Huan (Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University) ;
  • Wan, Jin-Nan (Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University) ;
  • Zhang, Yi-Min (Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University) ;
  • Tang, Zhao (Department of Pathology and Pathophysiology, Wuhan University School of Basic Medical Sciences) ;
  • Khan, Shahzad (Department of Pathology and Pathophysiology, Wuhan University School of Basic Medical Sciences) ;
  • Wang, Chang-Hua (Department of Pathology and Pathophysiology, Wuhan University School of Basic Medical Sciences) ;
  • Sun, Sheng-Rong (Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University)
  • Published : 2013.10.30

Abstract

Type 2 diabetes mellitus (T2DM) has contributed to advanced breast cancer development over the past decades. However, the mechanism underlying this contribution is poorly understood. In this study, we determined that high glucose enhanced proteasome activity was accompanied by enhanced proliferation, migration and invasion, as well as suppressed apoptosis, in human breast cancer MCF-7 cells. Proteasome inhibitor bortezomib (BZM) pretreatment mitigated high glucose-induced MCF-7 cell growth and invasion. Furthermore, high glucose increased protein kinase C delta ($PKC{\delta}$)-phosphorylation. Administration of the specific $PKC{\delta}$ inhibitor rottlerin attenuated high glucose-stimulated cancer cell growth and invasion. In addition, $PKC{\delta}$ inhibition by both rottlerin and $PKC{\delta}$ shRNA significantly suppressed high glucose-induced proteasome activity. Our results suggest that $PKC{\delta}$-dependent ubiquitin proteasome system activation plays an important role in high glucose-induced breast cancer cell growth and metastasis.

Keywords

References

  1. Bedford L, Lowe J, Dick LR, Mayer RJ, Brownell JE (2011). Ubiquitin-like protein conjugation and the ubiquitin-proteasome system as drug targets. Nat Rev Drug Discov, 10, 29-46. https://doi.org/10.1038/nrd3321
  2. Bence NF, Sampat RM, Kopito RR (2001). Impairment of the ubiquitin-proteasome system by protein aggregation. Science, 292, 1552-5. https://doi.org/10.1126/science.292.5521.1552
  3. Bugliani M, Liechti R, Cheon H, et al (2013). Microarray analysis of isolated human islet transcriptome in type 2 diabetes and the role of the ubiquitin-proteasome system in pancreatic beta cell dysfunction. Mol Cell Endocrinol, 367, 1-10. https://doi.org/10.1016/j.mce.2012.12.001
  4. Ciechanover A, Orian A, Schwartz AL (2000). Ubiquitin-mediated proteolysis: biological regulation via destruction. Bioessays, 22, 442-51. https://doi.org/10.1002/(SICI)1521-1878(200005)22:5<442::AID-BIES6>3.0.CO;2-Q
  5. Ciechanover A, Schwartz AL (1994). The ubiquitin-mediated proteolytic pathway: mechanisms of recognition of the proteolytic substrate and involvement in the degradation of native cellular proteins. FASEB J, 8, 182-91.
  6. Coughlin SS, Calle EE, Teras LR, Petrelli J, Thun MJ (2004). Diabetes mellitus as a predictor of cancer mortality in a large cohort of US adults. Am J Epidemiol, 159, 1160-7. https://doi.org/10.1093/aje/kwh161
  7. Cvek B, Dvorak Z (2011). The ubiquitin-proteasome system (UPS) and the mechanism of action of bortezomib. Curr Pharm Des, 17, 1483-99. https://doi.org/10.2174/138161211796197124
  8. Dang CV, Semenza GL (1999). Oncogenic alterations of metabolism. Trends Biochem Sci, 24, 68-72. https://doi.org/10.1016/S0968-0004(98)01344-9
  9. Dees EC, Orlowski RZ (2006). Targeting the ubiquitin-proteasome pathway in breast cancer therapy. Future Oncol, 2, 121-35. https://doi.org/10.2217/14796694.2.1.121
  10. Driscoll JJ, Woodle ES (2012). Targeting the ubiquitin+proteasome system in solid tumors. Semin Hematol, 49, 277-83. https://doi.org/10.1053/j.seminhematol.2012.04.002
  11. Gillies RJ, Gatenby RA (2007). Adaptive landscapes and emergent phenotypes: why do cancers have high glycolysis? J Bioenerg Biomembr, 39, 251-7. https://doi.org/10.1007/s10863-007-9085-y
  12. Goldberg AL (2003). Protein degradation and protection against misfolded or damaged proteins. Nature, 426, 895-9. https://doi.org/10.1038/nature02263
  13. Grossoni VC, Falbo KB, Kazanietz MG, de Kier Joffe ED, Urtreger AJ (2007). Protein kinase C delta enhances proliferation and survival of murine mammary cells. Mol Carcinog, 46, 381-90. https://doi.org/10.1002/mc.20287
  14. Hauptmann S, Grunewald V, Molls D, et al (2005). Glucose transporter GLUT1 in colorectal adenocarcinoma cell lines is inversely correlated with tumour cell proliferation. Anticancer Res, 25, 3431-6.
  15. Hochstrasser M (1995). Ubiquitin, proteasomes, and the regulation of intracellular protein degradation. Curr Opin Cell Biol, 7, 215-23. https://doi.org/10.1016/0955-0674(95)80031-X
  16. Jemal A, Bray F, Center MM, et al (2011). Global cancer statistics. CA Cancer J Clin, 61, 69-90. https://doi.org/10.3322/caac.20107
  17. Kiley SC, Clark KJ, Duddy SK, Welch DR, Jaken S (1999). Increased protein kinase C delta in mammary tumor cells: relationship to transformtion and metastatic progression. Oncogene, 18, 6748-57. https://doi.org/10.1038/sj.onc.1203101
  18. Kirk-Ballard H, Wang ZQ, Acharya P, et al (2013). An extract of Artemisia dracunculus L. inhibits ubiquitin-proteasome activity and preserves skeletal muscle mass in a murine model of diabetes. PLoS One, 8, e57112. https://doi.org/10.1371/journal.pone.0057112
  19. Kroemer G, Pouyssegur J (2008). Tumor cell metabolism: cancer's Achilles' heel. Cancer Cell, 13, 472-82. https://doi.org/10.1016/j.ccr.2008.05.005
  20. Landis-Piwowar KR, Milacic V, Chen D, et al (2006). The proteasome as a potential target for novel anticancer drugs and chemosensitizers. Drug Resist Updat, 9, 263-73. https://doi.org/10.1016/j.drup.2006.11.001
  21. Larsson SC, Mantzoros CS, Wolk A (2007). Diabetes mellitus and risk of breast cancer: a meta-analysis. Int J Cancer, 121, 856-62. https://doi.org/10.1002/ijc.22717
  22. Liao S, Li J, Wang L, et al (2010). Type 2 diabetes mellitus and characteristics of breast cancer in China. Asian Pac J Cancer Prev, 11, 933-7.
  23. Liao S, Li J, Wei W, et al (2011). Association between diabetes mellitus and breast cancer risk: a meta-analysis of the literature. Asian Pac J Cancer Prev, 12, 1061-5.
  24. Liu Z, Miers WR, Wei L, Barrett EJ (2000). The ubiquitin-proteasome proteolytic pathway in heart vs skeletal muscle: effects of acute diabetes. Biochem Biophys Res Commun, 276, 1255-60. https://doi.org/10.1006/bbrc.2000.3609
  25. Marfella R, Di Filippo C, D'Amico M, Paolisso G (2007). Diabetes, ubiquitin proteasome system and atherosclerotic plaque rupture. Circ Res, 100, e84-5. https://doi.org/10.1161/01.RES.0000269329.26803.25
  26. Marfella R, Di Filippo C, Portoghese M, et al (2009). The ubiquitin-proteasome system contributes to the inflammatory injury in ischemic diabetic myocardium: the role of glycemic control. Cardiovasc Pathol, 18, 332-45. https://doi.org/10.1016/j.carpath.2008.09.008
  27. Masur K, Vetter C, Hinz A, et al (2011). Diabetogenic glucose and insulin concentrations modulate transcriptome and protein levels involved in tumour cell migration, adhesion and proliferation. Br J Cancer, 104, 345-52. https://doi.org/10.1038/sj.bjc.6606050
  28. Micel LN, Tentler JJ, Smith PG, Eckhardt GS (2013). Role of ubiquitin ligases and the proteasome in oncogenesis: novel targets for anticancer therapies. J Clin Oncol, 31, 1231-8. https://doi.org/10.1200/JCO.2012.44.0958
  29. Naujokat C, Berges C, Hoh A, et al (2007). Proteasomal chymotrypsin-like peptidase activity is required for essential functions of human monocyte-derived dendritic cells. Immunology, 120, 120-32.
  30. Orlowski RZ, Dees EC (2003). The role of the ubiquitination-proteasome pathway in breast cancer: applying drugs that affect the ubiquitin-proteasome pathway to the therapy of breast cancer. Breast Cancer Res, 5, 1-7. https://doi.org/10.1186/bcr660
  31. Rubinsztein DC (2006). The roles of intracellular protein-degradation pathways in neurodegeneration. Nature, 443, 780-6. https://doi.org/10.1038/nature05291
  32. Sasaki S, Inoguchi T (2012). The role of oxidative stress in the pathogenesis of diabetic vascular complications. Diabetes Metab J, 36, 255-61. https://doi.org/10.4093/dmj.2012.36.4.255
  33. Sato K, Rajendra E, Ohta T (2008). The UPS: a promising target for breast cancer treatment. BMC Biochem, 9, S2. https://doi.org/10.1186/1471-2091-9-S1-S2
  34. Smith HJ, Wyke SM, Tisdale MJ (2004). Role of protein kinase C and NF-kappaB in proteolysis-inducing factor-induced proteasome expression in C(2)C(12) myotubes. Br J Cancer, 90, 1850-7.
  35. Srivastava AK (2002). High glucose-induced activation of protein kinase signaling pathways in vascular smooth muscle cells: a potential role in the pathogenesis of vascular dysfunction in diabetes (review). Int J Mol Med, 9, 85-9.
  36. Warburg, O (1956). On the origin of cancer cells. Science, 123, 309-14. https://doi.org/10.1126/science.123.3191.309
  37. Wing SS (2008). The UPS in diabetes and obesity. BMC Biochem, 9, S6. https://doi.org/10.1186/1471-2091-9-S1-S6
  38. Wolf I, Sadetzki S, Catane R, Karasik A, Kaufman B (2005). Diabetes mellitus and breast cancer. Lancet Oncol, 6, 103-11. https://doi.org/10.1016/S1470-2045(05)01736-5
  39. Wu WK, Cho CH, Lee CW, et al (2010). Proteasome inhibition: a new therapeutic strategy to cancer treatment. Cancer Lett, 293, 15-22. https://doi.org/10.1016/j.canlet.2009.12.002
  40. Wu WS (2006). The signaling mechanism of ROS in tumor progression. Cancer Metastasis Rev, 25, 695-705.

Cited by

  1. 20(S)-Protopanaxadiol Induces Human Breast Cancer MCF-7 Apoptosis through a Caspase-Mediated Pathway vol.15, pp.18, 2014, https://doi.org/10.7314/APJCP.2014.15.18.7919
  2. Cell Surface GRP78 Accelerated Breast Cancer Cell Proliferation and Migration by Activating STAT3 vol.10, pp.5, 2015, https://doi.org/10.1371/journal.pone.0125634
  3. UBE2Q1 in a Human Breast Carcinoma Cell Line: Overexpression and Interaction with p53 vol.16, pp.9, 2015, https://doi.org/10.7314/APJCP.2015.16.9.3723
  4. Curcumin Suppresses Proliferation and Migration of MDA-MB-231 Breast Cancer Cells through Autophagy-Dependent Akt Degradation vol.11, pp.1, 2016, https://doi.org/10.1371/journal.pone.0146553
  5. APPL1-Mediating Leptin Signaling Contributes to Proliferation and Migration of Cancer Cells vol.11, pp.11, 2016, https://doi.org/10.1371/journal.pone.0166172
  6. Epsilon-aminocaproic acid prevents high glucose and insulin induced-invasiveness in MDA-MB-231 breast cancer cells, modulating the plasminogen activator system pp.1573-4919, 2017, https://doi.org/10.1007/s11010-017-3096-8
  7. Induction of cell proliferation, clonogenicity and cell accumulation in S phase as a consequence of human UBE2Q1 overexpression vol.12, pp.3, 2016, https://doi.org/10.3892/ol.2016.4860
  8. Hyperglycemia and aberrant O-GlcNAcylation: contributions to tumor progression vol.50, pp.3, 2018, https://doi.org/10.1007/s10863-017-9740-x