Isaac Scientific Publishing

Journal of Advances in Molecular Biology

CaM Kinase I Regulation of p53 in Breast Cancer Cells

Download PDF (2973.1 KB) PP. 7 - 23 Pub. Date: June 1, 2020

DOI: 10.22606/jamb.2020.42001

Author(s)

  • Renee C. Geck
    The Biology Department, George Fox University, 414 N. Meridian St, Newberg, OR 97132, United States
  • Cody Coblentz
    The Biology Department, George Fox University, 414 N. Meridian St, Newberg, OR 97132, United States
  • Angela Rofelty
    The Biology Department, George Fox University, 414 N. Meridian St, Newberg, OR 97132, United States
  • John M. Schmitt*
    The Biology Department, George Fox University, 414 N. Meridian St, Newberg, OR 97132, United States

Abstract

Ca2+/calmodulin-dependent protein kinases (CaM Kinases) are intracellular signaling targets of hormones implicated in numerous cancers including breast cancer. The hormone estrogen (E2) or the muscarinic receptor agonist carbachol activate CaM Kinases, ERK and transcription factors to promote cell growth and differentiation. The mechanism by which E2 activates CaM Kinases and their precise intracellular targets that control cancer cell growth and survival remain to be elucidated. We show that E2 treatment of MCF-7 cells activates a CaM KI, ERK and cell growth pathway through the estrogen receptor alpha (ERα). Treatment of MDA-MB-231 (MDA) cells with carbachol also stimulated this pathway. Interestingly, we identified a CaM KI phosphorylation site within the C-terminal region of the tumor suppressor, p53, at serine 366 (S366). Stimulation of cells using either E2 or carbachol triggered phosphorylation of p53 at S366, which was blocked by pretreatment with the CaM KK inhibitor, STO-609 or transfection with siRNAs directed against CaM KI. Cells transfected with constitutively-active CaM KK revealed an elevated level of endogenous p53 phosphorylation at S366. Purified p53 associated with CaM KI from MCF-7 and MDA lysates and immunoprecipitation of transfected Flag-CaM KI revealed the presence of endogenous p53. Evaluation of agonist-stimulated cells using confocal microscopy suggests that endogenous CaM KI and p53 proteins co-localize in the cytoplasm. The p53 inhibitor, Pifithrin-alpha (PFT-α) increased p53 cytoplasmic localization, cell growth and survival. Treatment of MCF-7 or MDA cells with E2 or carbachol promoted cell growth and that was blocked by STO-609 but not PFT-α. This data is the first to suggest CaM KI may phosphorylate and regulate p53 in breast cancer cell growth and survival.

Keywords

Estrogen (E2), Carbachol, Calcium/calmodulin-dependent protein kinase kinase (CaM KK), Calcium/calmodulin-dependent protein kinase I (CaM KI), Extracellular-Regulated Protein Kinase (ERK), p53

References

[1] Brzozowski, J.S. and K.A. Skelding, The Multi-Functional Calcium/Calmodulin Stimulated Protein Kinase (CaMK) Family: Emerging Targets for Anti-Cancer Therapeutic Intervention. Pharmaceuticals (Basel), 2019. 12(1).

[2] Simon, B., A.S. Huart, and M. Wilmanns, Molecular mechanisms of protein kinase regulation by calcium/calmodulin. Bioorg Med Chem, 2015. 23(12): p. 2749-60.

[3] Takemoto-Kimura, S., et al., Calmodulin kinases: essential regulators in health and disease. J Neurochem, 2017. 141(6): p. 808-818.

[4] Bayer, K.U. and H. Schulman, CaM Kinase: Still Inspiring at 40. Neuron, 2019. 103(3): p. 380-394.

[5] Schmitt, J.M., et al., Calcium activation of ERK mediated by calmodulin kinase I. J Biol Chem, 2004. 279(23): p. 24064-72.

[6] Schmitt, J.M., et al., Calmodulin-dependent kinase kinase/calmodulin kinase I activity gates extracellularregulated kinase-dependent long-term potentiation. J Neurosci, 2005. 25(5): p. 1281-90.

[7] Schmitt, J.M., et al., ERK activation and cell growth require CaM kinases in MCF-7 breast cancer cells. Mol Cell Biochem, 2010. 335(1-2): p. 155-71.

[8] Kahl, C.R. and A.R. Means, Regulation of cyclin D1/Cdk4 complexes by calcium/calmodulin-dependent protein kinase I. J Biol Chem, 2004. 279(15): p. 15411-9.

[9] Lu, F., et al., Downregulation of CREB Promotes Cell Proliferation by Mediating G1/S Phase Transition in Hodgkin Lymphoma. Oncol Res, 2016. 24(3): p. 171-9.

[10] Sethi, S., et al., Social Context Enhances Hormonal Modulation of Pheromone Detection in Drosophila. Curr Biol, 2019. 29(22): p. 3887-3898 e4.

[11] Mallampalli, R.K., et al., Fbxl12 triggers G1 arrest by mediating degradation of calmodulin kinase I. Cell Signal, 2013.

[12] Kang, X., et al., CAMKs support development of acute myeloid leukemia. J Hematol Oncol, 2018. 11(1): p. 30.

[13] Davare, M.A., T. Saneyoshi, and T.R. Soderling, Calmodulin-kinases regulate basal and estrogen stimulated medulloblastoma migration via Rac1. J Neurooncol, 2011. 104(1): p. 65-82.

[14] Jadeja, R.N., et al., M3 muscarinic receptor activation reduces hepatocyte lipid accumulation via CaMKKbeta/AMPK pathway. Biochem Pharmacol, 2019. 169: p. 113613.

[15] Penfold, L., et al., CAMKK2 Promotes Prostate Cancer Independently of AMPK via Increased Lipogenesis. Cancer Res, 2018. 78(24): p. 6747-6761.

[16] Tokumitsu, H., et al., A single amino acid difference between alpha and beta Ca2+/calmodulin-dependent protein kinase kinase dictates sensitivity to the specific inhibitor, STO-609. J Biol Chem, 2003. 278(13): p. 10908-13.

[17] Rodriguez-Mora, O.G., et al., Calcium/calmodulin-dependent kinase I and calcium/calmodulin-dependent kinase kinase participate in the control of cell cycle progression in MCF-7 human breast cancer cells. Cancer Res, 2005. 65(12): p. 5408-16.

[18] Brufsky, A.M. and M.N. Dickler, Estrogen Receptor-Positive Breast Cancer: Exploiting Signaling Pathways Implicated in Endocrine Resistance. Oncologist, 2018. 23(5): p. 528-539.

[19] Jameera Begam, A., S. Jubie, and M.J. Nanjan, Estrogen receptor agonists/antagonists in breast cancer therapy: A critical review. Bioorg Chem, 2017. 71: p. 257-274.

[20] Improta-Brears, T., et al., Estrogen-induced activation of mitogen-activated protein kinase requires mobilization of intracellular calcium. Proc Natl Acad Sci U S A, 1999. 96(8): p. 4686-91.

[21] Pedram, A., et al., Developmental phenotype of a membrane only estrogen receptor alpha (MOER) mouse. J Biol Chem, 2009. 284(6): p. 3488-95.

[22] Bertheau, P., et al., p53 in breast cancer subtypes and new insights into response to chemotherapy. Breast, 2013. 22 Suppl 2: p. S27-9.

[23] Meek, D.W., Regulation of the p53 response and its relationship to cancer. Biochem J, 2015. 469(3): p. 325-46.

[24] Chen, J., The Cell-Cycle Arrest and Apoptotic Functions of p53 in Tumor Initiation and Progression. Cold Spring Harb Perspect Med, 2016. 6(3): p. a026104.

[25] Vousden, K.H. and C. Prives, Blinded by the Light: The Growing Complexity of p53. Cell, 2009. 137(3): p. 413- 31.

[26] Qu, L., et al., Endoplasmic reticulum stress induces p53 cytoplasmic localization and prevents p53-dependent apoptosis by a pathway involving glycogen synthase kinase-3beta. Genes Dev, 2004. 18(3): p. 261-77.

[27] Yogosawa, S. and K. Yoshida, Tumor suppressive role for kinases phosphorylating p53 in DNA damage-induced apoptosis. Cancer Sci, 2018. 109(11): p. 3376-3382.

[28] Rajagopalan, S., et al., 14-3-3 activation of DNA binding of p53 by enhancing its association into tetramers. Nucleic Acids Res, 2008. 36(18): p. 5983-91.

[29] Schmitt, J.M., et al., Estrogen Activation of CaM Kinases and Transcription Is Blocked by Vitamin D in MCF- 7 Breast Cancer Cells. Journal of Advances in Molecular Biology, 2017. 1(3): p. 129-147.

[30] Schmitt, J.M., et al., CaM kinase control of AKT and LNCaP cell survival. J Cell Biochem, 2012. 113(5): p. 1514-26.

[31] Gocher, A.M., et al., Akt activation by Ca(2+)/calmodulin-dependent protein kinase kinase 2 (CaMKK2) in ovarian cancer cells. J Biol Chem, 2017. 292(34): p. 14188-14204.

[32] Zhang, X., M.R. Diaz, and D. Yee, Fulvestrant regulates epidermal growth factor (EGF) family ligands to activate EGF receptor (EGFR) signaling in breast cancer cells. 2013. 139(2): p. 351-360.

[33] Chi, M., et al., Phosphorylation of calcium/calmodulin-stimulated protein kinase II at T286 enhances invasion and migration of human breast cancer cells. Sci Rep, 2016. 6: p. 33132.

[34] Foster, J.S., et al., Estrogens down-regulate p27Kip1 in breast cancer cells through Skp2 and through nuclear export mediated by the ERK pathway. J Biol Chem, 2003. 278(42): p. 41355-66.

[35] Fredersdorf, S., et al., High level expression of p27(kip1) and cyclin D1 in some human breast cancer cells: inverse correlation between the expression of p27(kip1) and degree of malignancy in human breast and colorectal cancers. Proc Natl Acad Sci U S A, 1997. 94(12): p. 6380-5.

[36] Li, W., et al., A truncated p53 in human lung cancer cells as a critical determinant of proliferation and invasiveness. Tumor Biology, 2017. 39(6): p. 101042831770382.

[37] Komarov, P.G., et al., A chemical inhibitor of p53 that protects mice from the side effects of cancer therapy. Science, 1999. 285(5434): p. 1733-7.

[38] Li, Q., et al., Genetic analysis of p53 nuclear importation. Oncogene, 2007. 26(57): p. 7885-93.

[39] Liang, S.H. and M.F. Clarke, A bipartite nuclear localization signal is required for p53 nuclear import regulated by a carboxyl-terminal domain. J Biol Chem, 1999. 274(46): p. 32699-703.

[40] Liang, S.H. and M.F. Clarke, The nuclear import of p53 is determined by the presence of a basic domain and its relative position to the nuclear localization signal. Oncogene, 1999. 18(12): p. 2163-6.

[41] Lin, F., et al., GTSE1 is involved in breast cancer progression in p53 mutation-dependent manner. Journal of Experimental & Clinical Cancer Research, 2019. 38(1).

[42] Korkmaz, G., et al., A CRISPR-Cas9 screen identifies essential CTCF anchor sites for estrogen receptor-driven breast cancer cell proliferation. Nucleic Acids Research, 2019. 47(18): p. 9557-9572.

[43] Bailey, S.T., et al., Estrogen receptor prevents p53-dependent apoptosis in breast cancer. Proc Natl Acad Sci U S A, 2012. 109(44): p. 18060-5.

[44] Ou, Y.H., et al., p53 C-terminal phosphorylation by CHK1 and CHK2 participates in the regulation of DNAdamage- induced C-terminal acetylation. Mol Biol Cell, 2005. 16(4): p. 1684-95.

[45] Rajagopalan, S., et al., Mechanistic differences in the transcriptional activation of p53 by 14-3-3 isoforms. Nucleic Acids Res, 2010. 38(3): p. 893-906.

[46] van Dieck, J., et al., Posttranslational modifications affect the interaction of S100 proteins with tumor suppressor p53. J Mol Biol, 2009. 394(5): p. 922-30.

[47] Mueller, A., et al., The calcium-binding protein S100A2 interacts with p53 and modulates its transcriptional activity. J Biol Chem, 2005. 280(32): p. 29186-93.