Protein kinase C (PKC) is a family of phospholipid-dependent serine/threonine kinases, which can be further classified into three PKC isozymes subfamilies: conventional or classic, novel or nonclassic, and atypical. PKC isozymes are known to be involved in cell proliferation, survival, invasion, migration, apoptosis, angiogenesis, and drug resistance. Because of their key roles in cell signaling, PKC isozymes also have the potential to be promising therapeutic targets for several diseases, such as cardiovascular diseases, immune and inflammatory diseases, neurological diseases, metabolic disorders, and multiple types of cancer. This review primarily focuses on the activation, mechanism, and function of PKC isozymes during cancer development and progression. 1. Introduction Protein kinase C (PKC) is a family of phospholipid-dependent serine/threonine kinases that function in numerous different cell types. Based on their structural and activation characteristics, this protein family can be further classified into three subfamilies: conventional or classic PKC isozymes (cPKCs; α, βI, βII, and γ), novel or nonclassic PKC isozymes (nPKCs; δ, ε, η, and θ), and atypical PKC isozymes (aPKCs; ζ, ι, and λ). The activation of cPKCs requires diacylglycerol (DAG) as the primary activator along with phosphatidylserine (PS) and calcium (Ca2+) as cofactors of activation. The nPKCs are also regulated by DAG and PS but do not require Ca2+ for activation. In the case of aPKCs, their activity is stimulated only by PS and not by DAG and Ca2+ [1, 2]. PKC isozymes are involved in multiple signal transduction systems that respond to a variety of external stimulators, including hormones, growth factors, and other membrane receptor ligands. For this reason, PKC isozymes can act as therapeutic targets for several diseases, such as cardiovascular diseases (e.g., atherosclerosis, myocardial fibrosis, cardiac hypertrophy, and hypertension) (for reviews, see [3, 4]), immune and inflammatory diseases (e.g., asthma, arthritis, and hepatitis) [5, 6], neurological diseases (e.g., Alzheimer’s disease and bipolar disorder) [7, 8], and metabolic disorders (e.g., obesity, insulin resistance, hyperglycemia, and hypercholesterolemia) [9–11]. Further, significant work has also explored the activation, mechanism, and function of PKC isozymes in the development and progression of multiple types of cancer, which will be the primary focus of this review. 2. PKC Isozymes and Their Target Proteins There are five consensus phosphorylation site motifs recognized by PKC isozymes, each of which has an
References
[1]
S. F. Steinberg, “Structural basis of protein kinase C isoform function,” Physiological Reviews, vol. 88, no. 4, pp. 1341–1378, 2008.
[2]
E. M. Griner and M. G. Kazanietz, “Protein kinase C and other diacylglycerol effectors in cancer,” Nature Reviews Cancer, vol. 7, no. 4, pp. 281–294, 2007.
[3]
E. Churchill, G. Budas, A. Vallentin, T. Koyanagi, and D. Mochly-Rosen, “PKC isozymes in chronic cardiac disease: possible therapeutic targets?” Annual Review of Pharmacology and Toxicology, vol. 48, pp. 569–599, 2008.
[4]
S. S. Palaniyandi, L. Sun, J. C. B. Ferreira, and D. Mochly-Rosen, “Protein kinase C in heart failure: a therapeutic target?” Cardiovascular Research, vol. 82, no. 2, pp. 229–239, 2009.
[5]
M. R. Lee, W. Duan, and S. Tan, “Protein kinase C isozymes as potential therapeutic targets in immune disorders,” Expert Opinion on Therapeutic Targets, vol. 12, no. 5, pp. 535–552, 2008.
[6]
D. H. Boschelli, “Small molecule inhibitors of PKCθ as potential antiinflammatory therapeutics,” Current Topics in Medicinal Chemistry, vol. 9, no. 7, pp. 640–654, 2009.
[7]
C. A. Zarate and H. K. Manji, “Protein kinase C inhibitors: rationale for use and potential in the treatment of bipolar disorder,” CNS Drugs, vol. 23, no. 7, pp. 569–582, 2009.
[8]
A. Pascale, M. Amadio, S. Govoni, and F. Battaini, “The aging brain, a key target for the future: the protein kinase C involvement,” Pharmacological Research, vol. 55, no. 6, pp. 560–569, 2007.
[9]
R. V. Farese and M. P. Sajan, “Atypical protein kinase C in cardiometabolic abnormalities,” Current Opinion in Lipidology, vol. 23, no. 3, pp. 175–181, 2012.
[10]
M. Clarke and P. M. Dodson, “PKC inhibition and diabetic microvascular complications,” Best Practice and Research: Clinical Endocrinology and Metabolism, vol. 21, no. 4, pp. 573–586, 2007.
[11]
N. Das Evcimen and G. L. King, “The role of protein kinase C activation and the vascular complications of diabetes,” Pharmacological Research, vol. 55, no. 6, pp. 498–510, 2007.
[12]
J. H.. Kang, R. Toita, C. W. Kim, and Y. Katayama, “Protein kinase C, (PKC) isozyme-specific substrates and their design,” Biotechnology Advances, vol. 30, no. 6, pp. 1662–1672, 2012.
[13]
H. J. Mackay and C. J. Twelves, “Targeting the protein kinase C family: are we there yet?” Nature Reviews Cancer, vol. 7, no. 7, pp. 554–562, 2007.
[14]
R. P. Danis and M. J. Sheetz, “Ruboxistaurin: PKC-β inhibition for complications of diabetes,” Expert Opinion on Pharmacotherapy, vol. 10, no. 17, pp. 2913–2925, 2009.
[15]
M. E. Sobhia, B. K. Grewal, S. P. Ml, et al., “Protein kinase C inhibitors: a patent review (2008-2009),” Expert Opinion on Therapeutic Patents, vol. 23, no. 10, pp. 1297–1315, 2013.
[16]
M. E. Sobhia, B. K. Grewal, M. L. Paul, et al., “Protein kinase C inhibitors: a patent review (2010—present),” Expert Opinion on Therapeutic Patents, vol. 23, no. 11, pp. 1451–1468, 2013.
[17]
K. Kielbassa, H.-J. Müller, H. E. Meyer, F. Marks, and M. Gschwendt, “Protein kinase Cδ-specific phosphorylation of the elongation factor eEF- 1α and an eEF-1α peptide at threonine 431,” The Journal of Biological Chemistry, vol. 270, no. 11, pp. 6156–6162, 1995.
[18]
J. H. Kang, D. Asai, S. Yamada et al., “A short peptide is a protein kinase C (PKC) α-specific substrate,” Proteomics, vol. 8, no. 10, pp. 2006–2011, 2008.
[19]
T. Kajimoto, S. Sawamura, Y. Tohyama, Y. Mori, and A. C. Newton, “Protein kinase C δ-specific activity reporter reveals agonist-evoked nuclear activity controlled by Src family of kinases,” The Journal of Biological Chemistry, vol. 285, no. 53, pp. 41896–41910, 2010.
[20]
Y. Serulle, G. Morfini, G. Pigino et al., “1-methyl-4-phenylpyridinium induces synaptic dysfunction through a pathway involving caspase and PKCδ enzymatic activities,” Proceedings of the National Academy of Sciences of the United States of America, vol. 104, no. 7, pp. 2437–2441, 2007.
[21]
V. O. Rybin, A. Sabri, J. Short, J. C. Braz, J. D. Molkentin, and S. F. Steinberg, “Cross-regulation of novel protein kinase C (PKC) isoform function in cardiomyocytes: role of PKCε in activation loop phosphorylations and PKCδ in hydrophobic motif phosphorylations,” The Journal of Biological Chemistry, vol. 278, no. 16, pp. 14555–14564, 2003.
[22]
J. H. Kang, D. Asai, R. Toita, H. Kitazaki, and Y. Katayama, “Plasma protein kinase C (PKC)α as a biomarker for the diagnosis of cancers,” Carcinogenesis, vol. 30, no. 11, pp. 1927–1931, 2009.
[23]
J. H. Kang, Y. Asami, M. Murata et al., “Gold nanoparticle-based colorimetric assay for cancer diagnosis,” Biosensors and Bioelectronics, vol. 25, no. 8, pp. 1869–1874, 2010.
[24]
J. H. Kang, J. Oishi, J. Kim et al., “Hepatoma-targeted gene delivery using a tumor cell-specific gene regulation system combined with a human liver cell-specific bionanocapsule,” Nanomedicine: Nanotechnology, Biology, and Medicine, vol. 6, no. 4, pp. 583–589, 2010.
[25]
J. H. Kang, D. Asai, J. Kim et al., “Design of polymeric carriers for cancer-specific gene targeting: utilization of abnormal protein kinase Cα activation in cancer cells,” Journal of the American Chemical Society, vol. 130, no. 45, pp. 14906–14907, 2008.
[26]
R. Toita, J. H.. Kang, T. Tomiyama, et al., “Gene carrier showing all-or-none response to cancer cell signaling,” Journal of the American Chemical Society, vol. 134, no. 37, pp. 15410–15417, 2012.
[27]
C.-C. Chen, J. Chang, and W.-C. Chen, “Role of protein kinase C subtypes α and δ in the regulation of bradykinin-stimulated phosphoinositide breakdown in astrocytes,” Molecular Pharmacology, vol. 48, no. 1, pp. 39–47, 1995.
[28]
A. Varga, G. Czifra, B. Tállai et al., “Tumor grade-dependent alterations in the protein kinase C isoform pattern in urinary bladder carcinomas,” European Urology, vol. 46, no. 4, pp. 462–465, 2004.
[29]
L. Langzam, R. Koren, R. Gal et al., “Patterns of protein kinase C isoenzyme expression in transitional cell carcinoma of bladder relation to degree of malignancy,” American Journal of Clinical Pathology, vol. 116, no. 3, pp. 377–385, 2001.
[30]
R. Koren, L. Langzam, A. Paz, P. M. Livne, R. Gal, and S. R. Sampson, “Protein kinase C (PKC) isoenzymes immunohistochemistry in lymph node revealing solution-fixed, paraffin-embedded bladder tumors,” Applied Immunohistochemistry and Molecular Morphology, vol. 8, no. 2, pp. 166–171, 2000.
[31]
C. Kong, Y. Zhu, D. Liu et al., “Role of protein kinase C-alpha in superficial bladder carcinoma recurrence,” Urology, vol. 65, no. 6, pp. 1228–1232, 2005.
[32]
A. V. Smrcka, J. H.. Brown, and G. G. Holz, “Role of phospholipase Cε in physiological phosphoinositide signaling networks,” Cellular Signalling, vol. 24, no. 6, pp. 1333–1343, 2012.
[33]
L. Yan, C. Luo, X. Wu, and Q. Zhang, “Involvement of the PLCε/PKCα pathway in human BIU-87 bladder cancer cell proliferation,” Cell Biology International, vol. 35, no. 10, pp. 1031–1036, 2011.
[34]
V. Aaltonen and J. Peltonen, “PKCα/β I inhibitor Go6976 induces dephosphorylation of constitutively hyperphosphorylated Rb and G1 arrest in T24 cells,” Anticancer Research, vol. 30, no. 10, pp. 3995–3999, 2010.
[35]
J. Koivunen, V. Aaltonen, S. Koskela, P. Lehenkar, M. Laato, and J. Peltonen, “Protein kinase C α/β inhibitor Go6976 promotes formation of cell junctions and inhibits invasion of urinary bladder carcinoma cells,” Cancer Research, vol. 64, no. 16, pp. 5693–5701, 2004.
[36]
K. A. Fitzgerald, A. G. Bowie, B. S. Skeffington, and L. A. J. O'Neill, “Ras, protein kinase Cζ, and IκB kinases 1 and 2 are downstream effectors of CD44 during the activation of NF-κB by hyaluronic acid fragments in T-24 carcinoma cells,” Journal of Immunology, vol. 164, no. 4, pp. 2053–2063, 2000.
[37]
M. Kim, H. Kwak, J. Lee et al., “17-allylamino-17-demethoxygeldanamycin down-regulates hyaluronic acid-induced glioma invasion by blocking matrix metalloproteinase-9 secretion,” Molecular Cancer Research, vol. 6, no. 11, pp. 1657–1665, 2008.
[38]
L. Y. W. Bourguignon, C. C. Spevak, G. Wong, W. Xia, and E. Gilad, “Hyaluronan-CD44 interaction with protein kinase Cε promotes oncogenic signaling by the stem cell marker nanog and the production of microRNA-21, leading to down-regulation of the tumor suppressor protein PDCD4, anti-apoptosis, and chemotherapy resistance in breast tumor cells,” The Journal of Biological Chemistry, vol. 284, no. 39, pp. 26533–26546, 2009.
[39]
C. Chen, P. Chan, S. Wang, Y. Pan, and H. Chen, “Elevated expression of protein kinase Cδ induces cell scattering upon serum deprivation,” Journal of Cell Science, vol. 123, no. 17, pp. 2901–2913, 2010.
[40]
A. K. Gupta, S. S. Galoforo, C. M. Berns et al., “Elevated levels of ERK2 in human breast carcinoma MCF-7 cells transfected with protein kinase Cα,” Cell Proliferation, vol. 29, no. 12, pp. 655–663, 1996.
[41]
H. Li, L. Zhao, Z. Yang, J. W. Funder, and J. Liu, “Telomerase is controlled by protein kinase Cα in human breast cancer cells,” The Journal of Biological Chemistry, vol. 273, no. 50, pp. 33436–33442, 1998.
[42]
J. Kim, S. H. Thorne, L. Sun, B. Huang, and D. Mochly-Rosen, “Sustained inhibition of PKCα reduces intravasation and lung seeding during mammary tumor metastasis in an in vivo mouse model,” Oncogene, vol. 30, no. 3, pp. 323–333, 2011.
[43]
S. Kim, J. Han, S. K. Lee et al., “Berberine suppresses the TPA-induced MMP-1 and MMP-9 expressions through the inhibition of PKC-α in breast cancer cells,” Journal of Surgical Research, vol. 176, no. 1, pp. e21–e29, 2012.
[44]
J. M. Connolly and D. P. Rose, “Expression of the invasive phenotype by MCF-7 human breast cancer cells transfected to overexpress protein kinase C-α or the erbB2 proto-oncogene,” International Journal of Oncology, vol. 10, no. 1, pp. 71–76, 1997.
[45]
M. Tan, P. Li, M. Sun, G. Yin, and D. Yu, “Upregulation and activation of PKCα by ErbB2 through Src promotes breast cancer cell invasion that can be blocked by combined treatment with PKCα and Src inhibitors,” Oncogene, vol. 25, no. 23, pp. 3286–3295, 2006.
[46]
J. D. Noti, “Adherence to osteopontin via alphavbeta3 suppresses phorbol ester-mediated apoptosis in MCF-7 breast cancer cells that overexpress protein kinase C-alpha,” International Journal of Oncology, vol. 17, no. 6, pp. 1237–1243, 2000.
[47]
D. B. Fournier, M. Chisamore, J. R. Lurain, A. W. Rademaker, V. C. Jordan, and D. A. Tonetti, “Protein kinase C alpha expression is inversely related to ER status in endometrial carcinoma: possible role in AP-1-mediated proliferation of ER-negative endometrial cancer,” Gynecologic Oncology, vol. 81, no. 3, pp. 366–372, 2001.
[48]
Z. Li, N. A. Wang, J. Fang, et al., “Role of PKC-ERK signaling in tamoxifen-induced apoptosis and tamoxifen resistance in human breast cancer cells,” Oncology Reports, vol. 27, no. 6, pp. 1879–1886, 2012.
[49]
J. W. Assender, J. M. W. Gee, I. Lewis, I. O. Ellis, J. F. R. Robertson, and R. I. Nicholson, “Protein kinase C isoform expression as a predictor of disease outcome on endocrine therapy in breast cancer,” Journal of Clinical Pathology, vol. 60, no. 11, pp. 1216–1221, 2007.
[50]
D. A. Tonetti, W. Gao, D. Escarzaga, K. Walter, A. Szafran, and J. S. Coon, “PKCα and ERβ are associated with triple-negative breast cancers in African American and Caucasian patients,” International Journal of Breast Cancer, vol. 2012, Article ID 740353, 9 pages, 2012.
[51]
W. L. Tam, H. Lu, J. Buikhuisen, et al., “Protein kinase C α is a central signaling node and therapeutic target for breast cancer stem cells,” Cancer Cell, vol. 24, no. 3, pp. 347–364, 2013.
[52]
D. A. Tonetti, M. Morrow, N. Kidwai, A. Gupta, and S. Badve, “Elevated protein kinase C alpha expression may be predictive of tamoxifen treatment failure,” British Journal of Cancer, vol. 88, no. 9, pp. 1400–1402, 2003.
[53]
L. B. Frankel, A. E. Lykkesfeldt, J. B. Hansen, and J. Stenvang, “Protein kinase C α is a marker for antiestrogen resistance and is involved in the growth of tamoxifen resistant human breast cancer cells,” Breast Cancer Research and Treatment, vol. 104, no. 2, pp. 165–179, 2007.
[54]
X. Lin, Y. Yu, H. Zhao, Y. Zhang, J. Manela, and D. A. Tonetti, “Overexpression of PKCα is required to impart estradiol inhibition and tamoxifen-resistance in a T47D human breast cancer tumor model,” Carcinogenesis, vol. 27, no. 8, pp. 1538–1546, 2006.
[55]
N. Wang, Z. Li, F. Tian et al., “PKCα inhibited apoptosis by decreasing the activity of JNK in MCF-7/ADR cells,” Experimental and Toxicologic Pathology, vol. 64, no. 5, pp. 459–464, 2012.
[56]
X.-F. Le, M. Marcelli, A. McWatters et al., “Heregulin-induced apoptosis is mediated by down-regulation of Bcl-2 and activation of caspase-7 and is potentiated by impairment of protein kinase C α activity,” Oncogene, vol. 20, no. 57, pp. 8258–8269, 2001.
[57]
P. K. Gill, A. Gescher, and T. W. Gant, “Regulation of MDR1 promoter activity in human breast carcinoma cells by protein kinase C isozymes α and θ,” European Journal of Biochemistry, vol. 268, no. 15, pp. 4151–4157, 2001.
[58]
S. Ahmad and R. I. Glazer, “Expression of the antisense cDNA for protein kinase Cα attenuates resistance in doxorubicin-resistant MCF-7 breast carcinoma cells,” Molecular Pharmacology, vol. 43, no. 6, pp. 858–862, 1993.
[59]
I. Carey, C. L. Williams, D. K. Ways, and J. D. M. Noti, “Overexpression of protein kinase C-α in MCF-7 breast cancer cells results in differential regulation and expression of αvβ3 and αvβ5,” International Journal of Oncology, vol. 15, no. 1, pp. 127–136, 1999.
[60]
G. K. L?nne, L. Cornmark, I. O. Zahirovic, G. Landberg, K. Jirstr?m, and C. Larsson, “PKCα expression is a marker for breast cancer aggressiveness,” Molecular Cancer, vol. 9, article 76, 2010.
[61]
H. Li and I. B. Weinstein, “Protein kinase C β enhances growth and expression of cyclin D1 in human breast cancer cells,” Cancer Research, vol. 66, no. 23, pp. 11399–11408, 2006.
[62]
V. C. Grossoni, L. B. Todaro, M. G. Kazanietz, E. D. Bal de Lier Joffé, and A. J. Urtreger, “Opposite effects of protein kinase C beta1 (PKCβ1) and PKCε in the metastatic potential of a breast cancer murine model,” Breast Cancer Research and Treatment, vol. 118, no. 3, pp. 469–480, 2009.
[63]
C. K?rner, I. Keklikoglou, C. Bender, A. W?rner, and E. Münstermann, “MicroRNA-31 sensitizes human breast cells to apoptosis by direct targeting of protein kinase C ? (PKC?),” The Journal of Biological Chemistry, vol. 288, no. 12, pp. 8750–8761, 2013.
[64]
M. H. Aziz, H. Shen, and C. G. Maki, “Glucocorticoid receptor activation inhibits p53-induced apoptosis of MCF10Amyc cells via induction of protein kinase Cε,” The Journal of Biological Chemistry, vol. 287, no. 35, pp. 29825–29836, 2012.
[65]
D. Lu, J. Huang, and A. Basu, “Protein kinase Cε activates protein kinase B/Akt via DNA-PK to protect against tumor necrosis factor-α-induced cell death,” The Journal of Biological Chemistry, vol. 281, no. 32, pp. 22799–22807, 2006.
[66]
E. Shankar, U. Sivaprasad, and A. Basu, “Protein kinase Cε confers resistance of MCF-7 cells to TRAIL by Akt-dependent activation of Hdm2 and downregulation of p53,” Oncogene, vol. 27, no. 28, pp. 3957–3966, 2008.
[67]
L. Huang, H. Cheng, R. Isom, C. Chen, R. A. Levine, and B. U. Pauli, “Protein kinase Cε mediates polymeric fibronectin assembly on the surface of blood-borne rat breast cancer cells to promote pulmonary metastasis,” The Journal of Biological Chemistry, vol. 283, no. 12, pp. 7616–7627, 2008.
[68]
D. Pal, S. P. Outram, and A. Basu, “Upregulation of PKCη by PKCε and PDK1 involves two distinct mechanisms and promotes breast cancer cell survival,” Biochimica et Biophysica Acta, vol. 1830, no. 8, pp. 4040–4045, 2013.
[69]
G. Karp, A. Maissel, and E. Livneh, “Hormonal regulation of PKC: estrogen up-regulates PKCη expression in estrogen-responsive breast cancer cells,” Cancer Letters, vol. 246, no. 1-2, pp. 173–181, 2007.
[70]
G. Karp, S. Abu-Ghanem, V. Novack, et al., “Localization of PKCη in cell membranes as a predictor for breast cancer response to treatment,” Onkologie, vol. 35, no. 5, pp. 260–266, 2012.
[71]
N. Rotem-Dai, G. Oberkovitz, S. Abu-Ghanem, and E. Livneh, “PKCη confers protection against apoptosis by inhibiting the pro-apoptotic JNK activity in MCF-7 cells,” Experimental Cell Research, vol. 315, no. 15, pp. 2616–2623, 2009.
[72]
G. R. Akkaraju and A. Basu, “Overexpression of protein kinase C-η attenuates caspase activation and tumor necrosis factor-α-induced cell death,” Biochemical and Biophysical Research Communications, vol. 279, no. 1, pp. 103–107, 2000.
[73]
P. A. Masso-Welch, J. S. Winston, S. Edge et al., “Altered expression and localization of PKC eta in human breast tumors,” Breast Cancer Research and Treatment, vol. 68, no. 3, pp. 211–223, 2001.
[74]
S. Huang, N. Ouyang, L. Lin et al., “HGF-induced PKCζ activation increases functional CXCR4 expression in human breast cancer cells,” PLoS ONE, vol. 7, no. 1, Article ID e29124, 2012.
[75]
P. Yi, Q. Feng, L. Amazit et al., “Atypical protein kinase C regulates dual pathways for degradation of the oncogenic coactivator SRC-3/ AIB1,” Molecular Cell, vol. 29, no. 4, pp. 465–476, 2008.
[76]
M. Mao, X. Fang, Y. Lu, R. LaPushin, R. C. Bast Jr., and G. B. Mills, “Inhibition of growth-factor-induced phosphorylation and activation of protein kinase B/Akt by atypical protein kinase C in breast cancer cells,” Biochemical Journal, vol. 352, no. 2, pp. 475–482, 2000.
[77]
K. Belguise, S. Milord, F. Galtier, G. Moquet-Torcy, M. Piechaczyk, and D. Chalbos, “The PKCθ pathway participates in the aberrant accumulation of Fra-1 protein in invasive ER-negative breast cancer cells,” Oncogene, vol. 31, no. 47, pp. 4889–4897, 2012.
[78]
K. Belguise and G. E. Sonenshein, “PKCθ promotes c-Rel-driven mammary tumorigenesis in mice and humans by repressing estrogen receptor α synthesis,” The Journal of Clinical Investigation, vol. 117, no. 12, pp. 4009–4021, 2007.
[79]
E. McKiernan, K. O'Brien, N. Grebenchtchikov et al., “Protein kinase Cδ expression in breast cancer as measured by real-time PCR, western blotting and ELISA,” British Journal of Cancer, vol. 99, no. 10, pp. 1644–1650, 2008.
[80]
S. Yin, S. Sethi, and K. B. Reddy, “Protein kinase Cδ and caspase-3 modulate TRAIL-induced apoptosis in breast tumor cells,” Journal of Cellular Biochemistry, vol. 111, no. 4, pp. 979–987, 2010.
[81]
J. Zhang, N. Liu, J. Zhang, S. Liu, Y. Liu, and D. Zheng, “PKCδ protects human breast tumor MCF-7 cells against tumor necrosis factor-related apoptosis-inducing ligand-mediated apoptosis,” Journal of Cellular Biochemistry, vol. 96, no. 3, pp. 522–532, 2005.
[82]
M. Kim, J. Ju, K. Jang et al., “Protein kinase Cδ negatively regulates Notch1-dependent transcription via a kinase-independent mechanism in vitro,” Biochimica et Biophysica Acta, vol. 1823, no. 2, pp. 387–397, 2012.
[83]
Y. S. Kim, H. T. An, J. Kim, and J. Ko, “Effects of protein kinase Cδ and phospholipase C-γ1 on monocyte chemoattractant protein-1 expression in taxol-induced breast cancer cell death,” International Journal of Molecular Medicine, vol. 24, no. 6, pp. 853–858, 2009.
[84]
M. I. Díaz Bessone, D. E. Berardi, P. B. Campodónico, et al., “Involvement of PKC delta (PKCδ) in the resistance against different doxorubicin analogs,” Breast Cancer Research and Treatment, vol. 126, no. 3, pp. 577–587, 2011.
[85]
V. C. Grossoni, K. B. Falbo, M. G. Kazanietz, E. D. Bal de Lier Joffé, and A. J. Urtreger, “Protein kinase C δ enhances proliferation and survival of murine mammary cells,” Molecular Carcinogenesis, vol. 46, no. 5, pp. 381–390, 2007.
[86]
S. M. Nabha, S. Glaros, M. Hong et al., “Upregulation of PKC-δ contributes to antiestrogen resistance in mammary tumor cells,” Oncogene, vol. 24, no. 19, pp. 3166–3176, 2005.
[87]
G. K. L?nne, K. C. Masoumi, J. Lennartsson, and C. Larsson, “Protein kinase Cδ supports survival of MDA-MB-231 breast cancer cells by suppressing the ERK1/2 pathway,” The Journal of Biological Chemistry, vol. 284, no. 48, pp. 33456–33465, 2009.
[88]
Y. H. Zeidan, B. X. Wu, R. W. Jenkins, L. M. Obeid, and Y. A. Hannun, “A novel role for protein kinase Cδ-mediated phosphorylation of acid sphingomyelinase in UV light-induced mitochondrial injury,” The FASEB Journal, vol. 22, no. 1, pp. 183–193, 2008.
[89]
S. A. Lee and M. Jung, “The nucleoside analog sangivamycin induces apoptotic cell death in breast carcinoma MCF7/adriamycin-resistant cells via protein kinase Cδ and JNK activation,” The Journal of Biological Chemistry, vol. 282, no. 20, pp. 15271–15283, 2007.
[90]
I. Vucenik, G. Ramakrishna, K. Tantivejkul, L. M. Anderson, and D. Ramljak, “Inositol hexaphosphate (IP6) blocks proliferation of human breast cancer cells through a PKCδ-dependent increase in p2 and decrease in retinoblastoma protein (pRb) phosphorylation,” Breast Cancer Research and Treatment, vol. 91, no. 1, pp. 35–45, 2005.
[91]
G. Yokoyama, T. Fujii, K. Tayama, H. Yamana, M. Kuwano, and K. Shirouzu, “PKCδ and MAPK mediate G1 arrest induced by PMA in SKBR-3 breast cancer cells,” Biochemical and Biophysical Research Communications, vol. 327, no. 3, pp. 720–726, 2005.
[92]
B. L. Allen-Petersen, C. J. Carter, A. M. Ohm, and M. E. Reyland, “Protein kinase Cδ is required for ErbB2-driven mammary gland tumorigenesis and negatively correlates with prognosis in human breast cancer,” Oncogene, vol. 33, no. 10, pp. 1306–1315, 2014.
[93]
S. Greco, C. Storelli, and S. Marsigliante, “Protein kinase C (PKC)-δ/-ε mediate the PKC/Akt-dependent phosphorylation of extracellular signal-regulated kinases 1 and 2 in MCF-7 cells stimulated by bradykinin,” Journal of Endocrinology, vol. 188, no. 1, pp. 79–89, 2006.
[94]
B. de Servi, A. Hermani, S. Medunjanin, and D. Mayer, “Impact of PKCδ on estrogen receptor localization and activity in breast cancer cells,” Oncogene, vol. 24, no. 31, pp. 4946–4955, 2005.
[95]
V. G. Keshamouni, R. R. Mattingly, and K. B. Reddy, “Mechanism of 17-β-estradiol-induced Erk1/2 activation in breast cancer cells. A role for HER2 and PKC-δ,” The Journal of Biological Chemistry, vol. 277, no. 25, pp. 22558–22565, 2002.
[96]
M. Shanmugam, N. L. Krett, E. T. Maizels, F. M. Murad, S. T. Rosen, and M. Hunzicker-Dunn, “A role for protein kinase C δ in the differential sensitivity of MCF-7 and MDA-MB 231 human breast cancer cells to phorbol ester-induced growth arrest and p2 induction,” Cancer Letters, vol. 172, no. 1, pp. 43–53, 2001.
[97]
Y. Zuo, Y. Wu, and C. Chakraborty, “Cdc42 negatively regulates intrinsic migration of highly aggressive breast cancer cells,” Journal of Cellular Physiology, vol. 227, no. 4, pp. 1399–1407, 2012.
[98]
C. Lin, W. Hou, S. Shen et al., “Quercetin inhibition of tumor invasion via suppressing PKCδ/ERK/ AP-1-dependent matrix metalloproteinase-9 activation in breast carcinoma cells,” Carcinogenesis, vol. 29, no. 9, pp. 1807–1815, 2008.
[99]
S. K. Park, Y. S. Hwang, K. Park, H. Park, J. Y. Seo, and W. Chung, “Kalopanaxsaponin A inhibits PMA-induced invasion by reducing matrix metalloproteinase-9 via PI3K/Akt- and PKCδ-mediated signaling in MCF-7 human breast cancer cells,” Carcinogenesis, vol. 30, no. 7, pp. 1225–1233, 2009.
[100]
D. Alonso-Escolano, C. Medina, K. Cieslik et al., “Protein kinase Cδ mediates platelet-induced breast cancer cell invasion,” Journal of Pharmacology and Experimental Therapeutics, vol. 318, no. 1, pp. 373–380, 2006.
[101]
D. Jackson, Y. Zheng, D. Lyo et al., “Suppression of cell migration by protein kinase Cδ,” Oncogene, vol. 24, no. 18, pp. 3067–3072, 2005.
[102]
L. A. Davidson, Y.-H. Jiang, J. N. Derr, H. M. Aukema, J. R. Lupton, and R. S. Chapkin, “Protein kinase C isoforms in human and rat colonic mucosa,” Archives of Biochemistry and Biophysics, vol. 312, no. 2, pp. 547–553, 1994.
[103]
S. Doi, D. Goldstein, H. Hug, and I. B. Weinstein, “Expression of multiple isoforms of protein kinase C in normal human colon mucosa and colon tumors and decreased levels of protein kinase C β and η mRNAs in the tumors,” Molecular Carcinogenesis, vol. 11, no. 4, pp. 197–203, 1994.
[104]
J. Pongracz, P. Clark, J. P. Neoptolemos, and J. M. Lord, “Expression of protein kinase C isoenzymes in colorectal cancer tissue and their differential activation by different bile acids,” International Journal of Cancer, vol. 61, no. 1, pp. 35–39, 1995.
[105]
G. Gobbi, D. di Marcantonio, C. Micheloni et al., “TRAIL up-regulation must be accompanied by a reciprocal PKCε down-regulation during differentiation of colonic epithelial cell: implications for colorectal cancer cell differentiation,” Journal of Cellular Physiology, vol. 227, no. 2, pp. 630–638, 2012.
[106]
I. Heider, B. Schulze, E. Oswald, P. Henklein, J. Scheele, and R. Kaufmann, “PAR1-type thrombin receptor stimulates migration and matrix adhesion of human colon carcinoma cells by a PKCε-dependent mechanism,” Oncology Research, vol. 14, no. 10, pp. 475–482, 2004.
[107]
M. L. Saxon, X. Zhao, and J. D. Black, “Activation of protein kinase C isozymes is associated with post-mitotic events in intestinal epithelial cells in situ,” The Journal of Cell Biology, vol. 126, no. 3, pp. 747–763, 1994.
[108]
G. Verstovsek, A. Byrd, M. R. Frey, N. J. Petrelli, and J. D. Black, “Colonocyte differentiation is associated with increased expression and altered distribution of protein kinase C isozymes,” Gastroenterology, vol. 115, no. 1, pp. 75–85, 1998.
[109]
B. Scaglione-Sewell, C. Abraham, M. Bissonnette et al., “Decreased PKC-α expression increases cellular proliferation, decreases differentiation, and enhances the transformed phenotype of CaCo-2 cells,” Cancer Research, vol. 58, no. 5, pp. 1074–1081, 1998.
[110]
H. Oster and M. Leitges, “Protein kinase C α but not PKCζ suppresses intestinal tumor formation in ApcMin/+ mice,” Cancer Research, vol. 66, no. 14, pp. 6955–6963, 2006.
[111]
M. A. Pysz, O. V. Leontieva, N. W. Bateman et al., “PKCα tumor suppression in the intestine is associated with transcriptional and translational inhibition of cyclin D1,” Experimental Cell Research, vol. 315, no. 8, pp. 1415–1428, 2009.
[112]
F. Hao, M. A. Pysz, K. J. Curry et al., “Protein kinase Cα signaling regulates inhibitor of DNA binding 1 in the intestinal epithelium,” The Journal of Biological Chemistry, vol. 286, no. 20, pp. 18104–18117, 2011.
[113]
J. Gwak, S. Jung, D. Kang et al., “Stimulation of protein kinase C-α suppresses colon cancer cell proliferation by down-regulation of β-catenin,” Journal of Cellular and Molecular Medicine, vol. 13, no. 8B, pp. 2171–2180, 2009.
[114]
J. M. Lee, I. S. Kim, H. Kim et al., “RORα attenuates Wnt/β-catenin signaling by PKCα-dependent phosphorylation in colon cancer,” Molecular Cell, vol. 37, no. 2, pp. 183–195, 2010.
[115]
B. Wu, H. Zhou, L. Hu, Y. Mu, and Y. Wu, “Involvement of PKCα activation in TF/VIIa/PAR2-induced proliferation, migration, and survival of colon cancer cell SW620,” Tumor Biology, vol. 34, no. 2, pp. 837–846, 2013.
[116]
K. Masur, K. Lang, B. Niggemann, K. S. Zanker, and F. Entschladen, “High PKC α and low E-cadherin expression contribute to high migratory activity of colon carcinoma cells,” Molecular Biology of the Cell, vol. 12, no. 7, pp. 1973–1982, 2001.
[117]
S. Chakrabarty, S. Rajagopal, and T. L. Moskal, “Protein kinase Cα controls the adhesion but not the antiproliferative response of human colon carcinoma cells to transforming growth factor β1: identification of two distinct branches of post-protein kinase Cα adhesion signal pathway,” Laboratory Investigation, vol. 78, no. 4, pp. 413–421, 1998.
[118]
H. Wang and S. Chakrabarty, “Requirement of protein kinase Cα, extracellular matrix remodeling, and cell-matrix interaction for transforming growth factorβ-regulated expression of E-cadherin and catenins,” Journal of Cellular Physiology, vol. 187, no. 2, pp. 188–195, 2001.
[119]
K. R. Gravitt, N. E. Ward, D. Fan, J. M. Skibber, B. Levin, and C. A. O'Brian, “Evidence that protein kinase C-α activation is a critical event in phorbol ester-induced multiple drug resistance in human colon cancer cells,” Biochemical Pharmacology, vol. 48, no. 2, pp. 375–381, 1994.
[120]
S. K. Lee, A. Shehza, J. C. Jung, et al., “Protein kinase Cα protects against multidrug resistance in human colon cancer cells,” Molecules and Cells, vol. 34, no. 1, pp. 61–69, 2012.
[121]
C. A. M. la Porta, E. Dolfini, and R. Comolli, “Inhibition of protein kinase C-α isoform enhances the P-glycoprotein expression and the survival of LoVo human colon adenocarcinoma cells to doxorubicin exposure,” British Journal of Cancer, vol. 78, no. 10, pp. 1283–1287, 1998.
[122]
S. Chakrabarty and S. Huang, “Modulation of chemosensitivity in human colon carcinoma cells by downregulating protein kinase Cα expression,” Journal of Experimental Therapeutics and Oncology, vol. 1, no. 4, pp. 218–221, 1996.
[123]
N. R. Murray, L. A. Davidson, R. S. Chapkin, W. C. Gustafson, D. G. Schattenberg, and A. P. Fields, “Overexpression of protein kinase C β(II) induces colonic hyperproliferation and increased sensitivity to colon carcinogenesis,” The Journal of Cell Biology, vol. 145, no. 4, pp. 699–711, 1999.
[124]
N. R. Murray, C. Weems, L. Chen et al., “Protein kinase C βII and TGFβRII in ω-3 fatty acid-mediated inhibition of colon carcinogenesis,” The Journal of Cell Biology, vol. 157, no. 6, pp. 915–920, 2002.
[125]
Y. G?kmen-Polar, N. R. Murray, M. A. Velasco, Z. Gatalica, and A. P. Fields, “Elevated protein kinase C βII is an early promotive event in colon carcinogenesis,” Cancer Research, vol. 61, no. 4, pp. 1375–1381, 2001.
[126]
Y. Liu, W. Su, E. A. Thompson, M. Leitges, N. R. Murray, and A. P. Fields, “Protein kinase C βII regulates its own expression in rat intestinal epithelial cells and the colonic epithelium in vivo,” The Journal of Biological Chemistry, vol. 279, no. 44, pp. 45556–45563, 2004.
[127]
W. Yu, N. R. Murray, C. Weems et al., “Role of cyclooxygenase 2 in protein kinase C βII-mediated colon carcinogenesis,” The Journal of Biological Chemistry, vol. 278, no. 13, pp. 11167–11174, 2003.
[128]
J. Zhang, P. Z. Anastasiadis, Y. Liu, E. A. Thompson, and A. P. Fields, “Protein kinase c (PKC) βII induces cell invasion through a Ras/Mek-, PKCι/Rac 1-dependent signaling pathway,” The Journal of Biological Chemistry, vol. 279, no. 21, pp. 22118–22123, 2004.
[129]
L. A. Davidson, R. E. Brown, W. L. Chang et al., “Morphodensitometric analysis of protein kinase C βII expression in rat colon: modulation by diet and relation to in situ cell proliferation and apoptosis,” Carcinogenesis, vol. 21, no. 8, pp. 1513–1519, 2000.
[130]
P. Cesaro, E. Raiteri, M. Démoz et al., “Expression of protein kinase C β1 confers resistance to TNFα- and paclitaxel-induced apoptosis in HT-29 colon carcinoma cells,” International Journal of Cancer, vol. 93, no. 2, pp. 179–184, 2001.
[131]
H. Lee, J. Ghose-Dastidar, S. Winawer, and E. Friedman, “Signal transduction through extracellular signal-regulated kinase-like pp57 blocked in differentiated cells having low protein kinase Cβ activity,” The Journal of Biological Chemistry, vol. 268, no. 7, pp. 5255–5263, 1993.
[132]
S. Sauma, Z. Yan, S. Ohno, and E. Friedman, “Protein kinase Cβ1 and protein kinase Cβ2 activate p57 mitogen- activated protein kinase and block differentiation in colon carcinoma cells,” Cell Growth & Differentiation, vol. 7, no. 5, pp. 587–594, 1996.
[133]
A. P. Fields, S. R. Calcagno, M. Krishna, S. Rak, M. Leitges, and N. R. Murray, “Protein kinase cβ is an effective target for chemoprevention of colon cancer,” Cancer Research, vol. 69, no. 4, pp. 1643–1650, 2009.
[134]
N. R. Murray, L. Jamieson, W. Yu et al., “Protein kinase Cι is required for Ras transformation and colon carcinogenesis in vivo,” The Journal of Cell Biology, vol. 164, no. 6, pp. 797–802, 2004.
[135]
N. R. Murray, J. Weems, U. Braun, M. Leitges, and A. P. Fields, “Protein kinase C βII and PKCι/λ: collaborating partners in colon cancer promotion and progression,” Cancer Research, vol. 69, no. 2, pp. 656–662, 2009.
[136]
S. I. Oikarinen, A. Pajari, I. Salminen, S. Heinonen, H. Adlercreutz, and M. Mutanen, “Effects of a flaxseed mixture and plant oils rich in α-linolenic acid on the adenoma formation in multiple intestinal neoplasia (Min) mice,” British Journal of Nutrition, vol. 94, no. 4, pp. 510–518, 2005.
[137]
R. Mustafi, S. Cerda, A. Chumsangsri, A. Fichera, and M. Bissonnette, “Protein kinase-ζ inhibits collagen I-dependent and anchorage-independent growth and enhances apoptosis of human Caco-2 cells,” Molecular Cancer Research, vol. 4, no. 9, pp. 683–694, 2006.
[138]
L. Ma, Y. Tao, A. Duran, et al., “Control of nutrient stress-induced metabolic reprogramming by PKCζ in tumorigenesis,” Cell, vol. 152, no. 3, pp. 599–611, 2013.
[139]
L. B. Luna-Ulloa, J. G. Hernández-Maqueda, P. Santoyo-Ramos, M. C. Casta?eda-Patlán, and M. Robles-Flores, “Protein kinase C ζ is a positive modulator of canonical wnt signaling pathway in tumoral colon cell lines,” Carcinogenesis, vol. 32, no. 11, pp. 1615–1624, 2011.
[140]
Y. Qin, Y. Tang, A. V. Schally, and B. S. Beckman, “Dexniguldipine hydrochloride inhibits growth of human HT-29 colon carcinoma cells and expression of protein kinase C Δ and ζ,” International Journal of Oncology, vol. 7, no. 5, pp. 1073–1077, 1995.
[141]
H. K. Roy, M. Bissonnette, B. P. Frawley Jr. et al., “Selective preservation of protein kinase C-ζ in the chemoprevention of azoxymethane-induced colonic tumors by piroxicam,” FEBS Letters, vol. 366, no. 2-3, pp. 143–145, 1995.
[142]
J. G. Hernández-Maqueda, L. B. Luna-Ulloa, P. Santoyo-Ramos, M. C. Casta?eda-Patlán, and M. Robles-Flores, “Protein kinase C delta negatively modulates canonical Wnt pathway and cell proliferation in colon tumor cell lines,” PLoS ONE, vol. 8, no. 3, Article ID e58540, 2013.
[143]
G. Perletti, E. Marras, D. Osti, L. Felici, S. Zaro, and M. de Eguileor, “PKCδ requires p53 for suppression of the transformed phenotype in human colon cancer cells,” Journal of Cellular and Molecular Medicine, vol. 8, no. 4, pp. 563–569, 2004.
[144]
G. Perletti, E. Marras, D. Dondi et al., “p2 and p53 are downstream effectors of protein kinase C delta in tumor suppression and differentiation in human colon cancer cells,” International Journal of Cancer, vol. 113, no. 1, pp. 42–53, 2005.
[145]
Y. H. Kim, J. H.. Lim, T. J. Lee, J. W. Park, and T. K. Kwon, “Expression of cyclin D3 through Sp1 sites by histone deacetylase inhibitors is mediated with protein kinase C-δ (PKC-δ) signal pathway,” Journal of Cellular Biochemistry, vol. 101, no. 4, pp. 987–995, 2007.
[146]
S. R. Cerda, R. Mustafi, H. Little et al., “Protein kinase C delta inhibits Caco-2 cell proliferation by selective changes in cell cycle and cell death regulators,” Oncogene, vol. 25, no. 22, pp. 3123–3138, 2006.
[147]
S. R. Cerda, M. Bissonnette, B. Scaglione-Sewell et al., “PKC-δ inhibits anchorage-dependent and -independent growth, enhances differentiation, and increases apoptosis in CaCo-2 cells,” Gastroenterology, vol. 120, no. 7, pp. 1700–1712, 2001.
[148]
A. E. Lewis, R. Susarla, B. C. Y. Wong, M. J. S. Langman, and M. C. Eggo, “Protein kinase C delta is not activated by caspase-3 and its inhibition is sufficient to induce apoptosis in the colon cancer line, COLO 205,” Cellular Signalling, vol. 17, no. 2, pp. 253–262, 2005.
[149]
Q. Wang, X. Wang, and B. M. Evers, “Induction of cIAP-2 in human colon cancer cells through PKCδ/NF-κB,” The Journal of Biological Chemistry, vol. 278, no. 51, pp. 51091–51099, 2003.
[150]
Q. Wang, X. Wang, Y. Zhou, and B. M. Evers, “PKCδ-mediated regulation of FLIP expression in human colon cancer cells,” International Journal of Cancer, vol. 118, no. 2, pp. 326–334, 2006.
[151]
D. H. Kho, J. A. Bae, J. H.. Lee et al., “KITENIN recruits Dishevelled/PKCd to form a functional complex and controls the migration and invasiveness of colorectal cancer cells,” Gut, vol. 58, no. 4, pp. 509–519, 2009.
[152]
M. Park, W. K. Kim, M. Song, et al., “Protein kinase C- -mediated recycling of active KIT in colon cancer,” Clinical Cancer Research, vol. 19, no. 18, pp. 4961–4971, 2013.
[153]
A. Doller, S. Schulz, J. Pfeilschifter, and W. Eberhardt, “RNA-dependent association with myosin IIA promotes F-actin-guided trafficking of the ELAV-like protein HuR to polysomes,” Nucleic Acids Research, vol. 41, no. 19, pp. 9152–9167, 2013.
[154]
A. Sadok, V. Bourgarel-Rey, F. Gattacceca, C. Penel, M. Lehmann, and H. Kovacic, “Nox1-dependent superoxide production controls colon adenocarcinoma cell migration,” Biochimica et Biophysica Acta, vol. 1783, no. 1, pp. 23–33, 2008.
[155]
S. C. Lin, W. Y. Chen, K. Y. Lin, et al., “Clinicopathological correlation and prognostic significance of protein kinase Cα overexpression in human gastric carcinoma,” PLoS ONE, vol. 8, no. 2, Article ID e56675, 2013.
[156]
K. Lin, C. Fang, Y. Uen et al., “Overexpression of protein kinase Cα mRNA may be an independent prognostic marker for gastric carcinoma,” Journal of Surgical Oncology, vol. 97, no. 6, pp. 538–543, 2008.
[157]
Y. Han, Z. Han, X. Zhou et al., “Expression and function of classical protein kinase C isoenzymes in gastric cancer cell line and its drug-resistant sublines,” World Journal of Gastroenterology, vol. 8, no. 3, pp. 441–445, 2002.
[158]
D. Wu, F. Sui, C. Du et al., “Antisense expression of PKCα improved sensitivity of SGC7901/VCR cells to doxorubicin,” World Journal of Gastroenterology, vol. 15, no. 10, pp. 1259–1263, 2009.
[159]
X. Jiang, S. Tu, J. Cui et al., “Antisense targeting protein kinase C α and β1 inhibits gastric carcinogenesis,” Cancer Research, vol. 64, no. 16, pp. 5787–5794, 2004.
[160]
M. J. Redlak, J. J. Power, and T. A. Miller, “Aspirin-induced apoptosis in human gastric cancer epithelial cells: relationship with protein kinase C signaling,” Digestive Diseases and Sciences, vol. 52, no. 3, pp. 810–816, 2007.
[161]
M. J. Atten, E. Godoy-Romero, B. M. Attar, T. Milson, M. Zopel, and O. Holian, “Resveratrol regulates cellular PKC α and δ to inhibit growth and induce apoptosis in gastric cancer cells,” Investigational New Drugs, vol. 23, no. 2, pp. 111–119, 2005.
[162]
H. Okuda, M. Adachi, M. Miyazawa, Y. Hinoda, and K. Imai, “Protein kinase Cα promotes apoptotic cell death in gastric cancer cells depending upon loss of anchorage,” Oncogene, vol. 18, no. 40, pp. 5604–5609, 1999.
[163]
B. Zhang and C. Xia, “12-O-tetradecanoylphorbol-1,3-acetate induces the negative regulation of protein kinase B by protein kinase Cα during gastric cancer cell apoptosis,” Cellular and Molecular Biology Letters, vol. 15, no. 3, pp. 377–394, 2010.
[164]
G. H. Zhu, B. C. Y. Wong, E. D. Slosberg et al., “Overexpression of protein kinase C-β1 isoenzyme suppresses indomethacin-induced apoptosis in gastric epithelial cells,” Gastroenterology, vol. 118, no. 3, pp. 507–514, 2000.
[165]
X. Jiang, S. Lam, M. C. M. Lin et al., “Novel target for induction of apoptosis by cyclo-oxygenase-2 inhibitor SC-236 through a protein kinase C-β1-dependent pathway,” Oncogene, vol. 21, no. 39, pp. 6113–6122, 2002.
[166]
K. W. Lee, S. G. Kim, H. P. Kim, et al., “Enzastaurin, a protein kinase Cβ inhibitor, suppresses signaling through the ribosomal S6 kinase and bad pathways and induces apoptosis in human gastric cancer cells,” Cancer Research, vol. 68, no. 6, pp. 1916–1926, 2008.
[167]
M. Lisovsky, F. Ogawa, K. Dresser, B. Woda, and G. Y. Lauwers, “Loss of cell polarity protein Lgl2 in foveolar-type gastric dysplasia: correlation with expression of the apical marker aPKC-zeta,” Virchows Archiv, vol. 457, no. 6, pp. 635–642, 2010.
[168]
R. Takagawa, K. Akimoto, Y. Ichikawa et al., “High expression of atypical protein kinase C λ/ι in gastric cancer as a prognostic factor for recurrence,” Annals of Surgical Oncology, vol. 17, no. 1, pp. 81–88, 2010.
[169]
Y. Yoshihama, Y. Izumisawa, K. Akimoto, et al., “High expression of KIBRA in low atypical protein kinase C-expressing gastric cancer correlates with lymphatic invasion and poor prognosis,” Cancer Science, vol. 104, no. 2, pp. 259–265, 2013.
[170]
Y. Iioka, K. Mishima, N. Azuma et al., “Overexpression of protein kinase Cδ enhances cisplatin-induced cytotoxicity correlated with p53 in gastric cancer cell line,” Pathobiology, vol. 72, no. 3, pp. 152–159, 2005.
[171]
T. Kanno, T. Nishimoto, Y. Fujita, A. Gotoh, T. Nakano, and T. Nishizaki, “Sphingosine induces apoptosis in MKN-28 human gastric cancer cells in an SDK-dependent manner,” Cellular Physiology and Biochemistry, vol. 30, pp. 987–994, 2012.
[172]
M. Lee, T. Y. Kim, Y. Kim et al., “The signaling network of transforming growth factor β1, protein kinase Cδ and integrin underlies the spreading and invasiveness of gastric carcinoma cells,” Molecular and Cellular Biology, vol. 25, no. 16, pp. 6921–6936, 2005.
[173]
H. E. Lee, M. A. Kim, H. S. Lee, B. L. Lee, and W. H. Kim, “Characteristics of KIT-negative gastrointestinal stromal tumours and diagnostic utility of protein kinase C theta immunostaining,” Journal of Clinical Pathology, vol. 61, no. 6, pp. 722–729, 2008.
[174]
P. Blay, A. Astudillo, J. M. Buesa et al., “Protein kinase C θ is highly expressed in gastrointestinal stromal tumors but not in other mesenchymal neoplasias,” Clinical Cancer Research, vol. 10, no. 12, part 1, pp. 4089–4095, 2004.
[175]
W.-B. Ou, M.-J. Zhu, G. D. Demetri, C. D. M. Fletcher, and J. A. Fletcher, “Protein kinase C-θ regulates KIT expression and proliferation in gastrointestinal stromal tumors,” Oncogene, vol. 27, no. 42, pp. 5624–5634, 2008.
[176]
A. Motegi, S. Sakurai, H. Nakayama, T. Sano, T. Oyama, and T. Nakajima, “PKC theta, a novel immunohistochemical marker for gastrointestinal stromal tumors (GIST), especially useful for identifying KIT-negative tumors,” Pathology International, vol. 55, no. 3, pp. 106–112, 2005.
[177]
G. Kang, A. Srivastava, Y. E. Kim et al., “DOG1 and PKC-θ-are useful in the diagnosis of KIT-negative gastrointestinal stromal tumors,” Modern Pathology, vol. 24, no. 6, pp. 866–875, 2011.
[178]
C. Wang, M. S. Jin, Y. B. Zou, et al., “Diagnostic significance of DOG-1 and PKC-θ expression and c-Kit/PDGFRA mutations in gastrointestinal stromal tumours,” Scandinavian Journal of Gastroenterology, vol. 48, no. 9, pp. 1055–1065, 2013.
[179]
K. Kim, D. W. Kang, W. S. Moon et al., “PKCθ expression in gastrointestinal stromal tumor,” Modern Pathology, vol. 19, no. 11, pp. 1480–1486, 2006.
[180]
M. J. Ríos-Moreno, S. Jaramillo, S. Pereira Gallardo et al., “Gastrointestinal stromal tumors (GISTs): CD117, DOG-1 and PKCθ expression. Is there any advantage in using several markers?” Pathology Research and Practice, vol. 208, no. 2, pp. 74–81, 2012.
[181]
P. Y. Wen and S. Kesari, “Malignant gliomas in adults,” The New England Journal of Medicine, vol. 359, no. 5, pp. 492–507, 2008.
[182]
C.-C. Chen, “Protein kinase Cα, δ, ε and ζ in C6 glioma cells. TPA induces translocation and down-regulation of conventional and new PKC isoforms but not atypical PKC ζ,” FEBS Letters, vol. 332, no. 1-2, pp. 169–173, 1993.
[183]
M. I. González, B. T. S. Susarla, and M. B. Robinson, “Evidence that protein kinase Cα interacts with and regulates the glial glutamate transporter GLT-1,” Journal of Neurochemistry, vol. 94, no. 5, pp. 1180–1188, 2005.
[184]
H. Xiao, D. A. Goldthwait, and T. Mapstone, “The identification of four protein kinase C isoforms in human glioblastoma cell lines: PKC alpha, gamma, epsilon, and zeta,” Journal of Neurosurgery, vol. 81, no. 5, pp. 734–740, 1994.
[185]
A. Misra-Press, A. P. Fields, D. Samols, and D. A. Goldthwait, “Protein kinase C isoforms in human glioblastoma cells,” Glia, vol. 6, no. 3, pp. 188–197, 1992.
[186]
R. Mandil, E. Ashkenazi, M. Blass et al., “Protein kinase Cα and protein kinase Cδ play opposite roles in the proliferation and apoptosis of glioma cells,” Cancer Research, vol. 61, no. 11, pp. 4612–4619, 2001.
[187]
T. Todo, N. Shitara, H. Nakamura, K. Takakura, and K. Ikeda, “Immunohistochemical demonstration of protein kinase C isozymes in human brain tumors,” Neurosurgery, vol. 29, no. 3, pp. 399–404, 1991.
[188]
D. L. Benzil, S. D. Finkelstein, M. H. Epstein, and P. W. Finch, “Expression pattern of α-protein kinase C in human astrocytomas indicates a role in malignant progression,” Cancer Research, vol. 52, no. 10, pp. 2951–2956, 1992.
[189]
M. Leirdal and M. Sioud, “Protein kinase Cα isoform regulates the activation of the MAP kinase ERK1/2 in human glioma cells: involvement in cell survival and gene expression,” Molecular Cell Biology Research Communications, vol. 4, no. 2, pp. 106–110, 2000.
[190]
Q. Fan, C. Cheng, Z. A. Knight et al., “EGFR signals to mTOR through PKC and independently of Akt in glioma,” Science Signaling, vol. 2, no. 55, article ra4, 2009.
[191]
A. J. Cameron, K. J. Procyk, M. Leitges, and P. J. Parker, “PKC alpha protein but not kinase activity is critical for glioma cell proliferation and survival,” International Journal of Cancer, vol. 123, no. 4, pp. 769–779, 2008.
[192]
R. Sattler, B. Tyler, B. Hoover, et al., “Increased expression of glutamate transporter GLT-1 in peritumoral tissue associated with prolonged survival and decreases in tumor growth in a rat model of experimental malignant glioma,” Journal of Neurosurgery, vol. 119, no. 4, pp. 878–886, 2013.
[193]
M. I. González, P. G. Bannerman, and M. B. Robinson, “Phorbol myristate acetate-dependent interaction of protein kinase Cα and the neuronal glutamate transporter EAAC1,” Journal of Neuroscience, vol. 23, no. 13, pp. 5589–5593, 2003.
[194]
Y. Huang and Z. Zuo, “Isoflurane induces a protein kinase C α-dependent increase in cell-surface protein level and activity of glutamate transporter type 3,” Molecular Pharmacology, vol. 67, no. 5, pp. 1522–1533, 2005.
[195]
A. M. Donson, A. Banerjee, F. Gamboni-Robertson, J. M. Fleitz, and N. K. Foreman, “Protein kinase C ζ isoform is critical for proliferation in human glioblastoma cell lines,” Journal of Neuro-Oncology, vol. 47, no. 2, pp. 109–115, 2000.
[196]
S. Singh, T. Okamura, and F. Ali-Osman, “Serine phosphorylation of glutathione S-transferase P1 (GSTP1) by PKCα enhances GSTP1-dependent cisplatin metabolism and resistance in human glioma cells,” Biochemical Pharmacology, vol. 80, no. 9, pp. 1343–1355, 2010.
[197]
D. R. Sorensen, M. Leirdal, P. O. Iversen, and M. Sioud, “Combination of endostatin and a protein kinase Cα DNA enzyme improves the survival of rats with malignant glioma,” Neoplasia, vol. 4, no. 6, pp. 474–479, 2002.
[198]
M. Leirdal and M. Sioud, “Ribozyme inhibition of the protein kinase Cα triggers apoptosis in glioma cells,” British Journal of Cancer, vol. 80, no. 10, pp. 1558–1564, 1999.
[199]
S. A. Grossman, J. B. Alavi, J. G. Supko et al., “Efficacy and toxicity of the antisense oligonucleotide aprinocarsen directed against protein kinase C-α delivered as a 21-day continuous intravenous infusion in patients with recurrent high-grade astrocytomas,” Neuro-Oncology, vol. 7, no. 1, pp. 32–40, 2005.
[200]
C. Lin, S. Shen, C. Chien, L. Yang, L. Shia, and Y. Chen, “12-O-tetradecanoylphorbol-13-acetate-induced invasion/migration of glioblastoma cells through activating PKCα/ERK/NF-κB-dependent MMP-9 expression,” Journal of Cellular Physiology, vol. 225, no. 2, pp. 472–481, 2010.
[201]
S. Amos, M. Mut, C. G. diPierro et al., “Protein kinase C-α-mediated regulation of low-density lipoprotein receptor-related protein and urokinase increases astrocytoma invasion,” Cancer Research, vol. 67, no. 21, pp. 10241–10251, 2007.
[202]
Z. A. Kohutek, C. G. diPierro, G. T. Redpath, and I. M. Hussaini, “ADAM-10-mediated N-cadherin cleavage is protein kinase C-α dependent and promotes glioblastoma cell migration,” Journal of Neuroscience, vol. 29, no. 14, pp. 4605–4615, 2009.
[203]
I. T. Makagiansar, S. Williams, T. Mustelin, and W. B. Stallcup, “Differential phosphorylation of NG2 proteoglycan by ERK and PKCα helps balance cell proliferation and migration,” The Journal of Cell Biology, vol. 178, no. 1, pp. 155–165, 2007.
[204]
A. Ziv-Av, D. Taller, M. Attia et al., “RTVP-1 expression is regulated by SRF downstream of protein kinase C and contributes to the effect of SRF on glioma cell migration,” Cellular Signalling, vol. 23, no. 12, pp. 1936–1943, 2011.
[205]
T. R. Sharif and M. Sharif, “Overexpression of protein kinase C epsilon in astroglial brain tumor derived cell lines and primary tumor samples,” International Journal of Oncology, vol. 15, no. 2, pp. 237–243, 1999.
[206]
S. Kahana, S. Finniss, S. Cazacu et al., “Proteasome inhibitors sensitize glioma cells and glioma stem cells to TRAIL-induced apoptosis by PKCε-dependent downregulation of AKT and XIAP expressions,” Cellular Signalling, vol. 23, no. 8, pp. 1348–1357, 2011.
[207]
H. Shinohara, N. Kayagaki, H. Yagita et al., “A protective role of PKCε against TNF-related apoptosis-inducing ligand (TRAIL)-induced apoptosis in glioma cells,” Biochemical and Biophysical Research Communications, vol. 284, no. 5, pp. 1162–1167, 2001.
[208]
M. H. Aziz, B. B. Hafeez, J. M. Sand et al., “Protein kinase Cε mediates Stat3Ser727 phosphorylation, Stat3-regulated gene expression, and cell invasion in various human cancer cell lines through integration with MAPK cascade (RAF-1, MEK1/2, and ERK1/2),” Oncogene, vol. 29, no. 21, pp. 3100–3109, 2010.
[209]
S. Fortin, M. le Mercier, I. Camby et al., “Galectin-1 is implicated in the protein kinase c ε/vimentin-controlled trafficking of integrin-β1 in glioblastoma cells,” Brain Pathology, vol. 20, no. 1, pp. 39–49, 2010.
[210]
A. Besson, A. Davy, S. M. Robbins, and V. W. Yong, “Differential activation of ERKs to focal adhesions by PKC ε is required for PMA-induced adhesion and migration of human glioma cells,” Oncogene, vol. 20, no. 50, pp. 7398–7407, 2001.
[211]
T. R. Sharif, N. Sasakawa, and M. Sharif, “Regulated expression of a dominant negative protein kinase C epsilon mutant inhibits the proliferation of U-373MG human astrocytoma cells,” International Journal of Molecular Medicine, vol. 7, no. 4, pp. 373–380, 2001.
[212]
K. Alapati, S. Gopinath, R. Malla, V. R. Dasari, and J. Rao, “uPAR and cathepsin B knockdown inhibits radiation-induced PKC integrated integrin signaling to the cytoskeleton of glioma-initiating cells,” International Journal of Oncology, vol. 41, no. 2, pp. 599–610, 2012.
[213]
H. Guo, F. Gu, W. Li et al., “Reduction of protein kinase C ζ inhibits migration and invasion of human glioblastoma cells,” Journal of Neurochemistry, vol. 109, no. 1, pp. 203–213, 2009.
[214]
H. Huang, C. Huang, S. Lin-Shiau, and J. Lin, “Ursolic acid inhibits IL-1β or TNF-α-induced C6 glioma invasion through suppressing the association ZIP/p62 with PKC-ζ and downregulating the MMP-9 expression,” Molecular Carcinogenesis, vol. 48, no. 6, pp. 517–531, 2009.
[215]
P. O. Estève, é. Chicoine, O. Robledo et al., “Protein kinase C-ζ regulates transcription of the matrix metalloproteinase-9 gene induced by IL-1 and TNF-α in glioma cells via NF-κB,” The Journal of Biological Chemistry, vol. 277, no. 38, pp. 35150–35155, 2002.
[216]
K. van Kolen and H. Slegers, “Atypical PKCζ is involved in RhoA-dependent mitogenic signaling by the P2Y12 receptor in C6 cells,” The FEBS Journal, vol. 273, no. 8, pp. 1843–1854, 2006.
[217]
M. Guizzetti and L. G. Costa, “Effect of ethanol on protein kinase Cζ and p70S6 kinase activation by carbachol: a possible mechanism for ethanol-induced inhibition of glial cell proliferation,” Journal of Neurochemistry, vol. 82, no. 1, pp. 38–46, 2002.
[218]
R. Patel, H. Win, S. Desai, K. Patel, J. A. Matthews, and M. Acevedo-Duncan, “Involvement of PKC-ι in glioma proliferation,” Cell Proliferation, vol. 41, no. 1, pp. 122–135, 2008.
[219]
E. Bicaku, R. Patel, and M. Acevedo-Duncan, “Cyclin-dependent kinase activating kinase/Cdk7 co-localizes with PKC-ι in human glioma cells,” Tissue and Cell, vol. 37, no. 1, pp. 53–58, 2005.
[220]
S. R. Desai, P. P. Pillai, R. S. Patel, A. N. McCray, H. Y. Win-Piazza, and M. E. Acevedo-Duncan, “Regulation of Cdk7 activity through a phosphatidylinositol (3)-kinase/PKC-ι-mediated signaling cascade in glioblastoma,” Carcinogenesis, vol. 33, no. 1, pp. 10–19, 2012.
[221]
S. Desai, P. Pillai, H. Win-Piazza, and M. Acevedo-Duncan, “PKC-3 promotes glioblastoma cell survival by phosphorylating and inhibiting BAD through a phosphatidylinositol 3-kinase pathway,” Biochimica et Biophysica Acta, vol. 1813, no. 6, pp. 1190–1197, 2011.
[222]
R. M. Baldwin, M. Garratt-Lalonde, D. A. E. Parolin, P. M. Krzyzanowski, M. A. Andrade, and I. A. J. Lorimer, “Protection of glioblastoma cells from cisplatin cytotoxicity via protein kinase Cι-mediated attenuation of p38 MAP kinase signaling,” Oncogene, vol. 25, no. 20, pp. 2909–2919, 2006.
[223]
R. M. Baldwin, D. A. E. Parolin, and I. A. J. Lorimer, “Regulation of glioblastoma cell invasion by PKCι and RhoB,” Oncogene, vol. 27, no. 25, pp. 3587–3595, 2008.
[224]
R. M. Uht, S. Amos, P. M. Martin, A. E. Riggan, and I. M. Hussaini, “The protein kinase C-η isoform induces proliferation in glioblastoma cell lines through an ERK/Elk-1 pathway,” Oncogene, vol. 26, no. 20, pp. 2885–2893, 2007.
[225]
S. E. Aeder, P. M. Martin, J. Soh, and I. M. Hussaini, “PKC-η mediates glioblastoma cell proliferation through the Akt and mTOR signaling pathways,” Oncogene, vol. 23, no. 56, pp. 9062–9069, 2004.
[226]
P. M. Martin, S. E. Aeder, C. A. Chrestensen, T. W. Sturgill, and I. M. Hussaini, “Phorbol 12-myristate 13-acetate and serum synergize to promote rapamycin-insensitive cell proliferation via protein kinase C-eta,” Oncogene, vol. 26, no. 3, pp. 407–414, 2007.
[227]
I. M. Hussaini, J. E. Carpenter, G. T. Redpath, J. J. Sando, M. E. Shaffrey, and S. R. VandenBerg, “Protein kinase C-η regulates resistance to UV- and γ-irradiation-induced apoptosis in glioblastoma cells by preventing caspase-9 activation,” Neuro-Oncology, vol. 4, no. 1, pp. 9–21, 2002.
[228]
W. Lu, H. Lee, C. Xiang, S. Finniss, and C. Brodie, “The phosphorylation of tyrosine 332 is necessary for the caspase 3-dependent cleavage of PKCδ and the regulation of cell apoptosis,” Cellular Signalling, vol. 19, no. 10, pp. 2165–2173, 2007.
[229]
W. Lu, S. Finnis, C. Xiang et al., “Tyrosine 311 is phosphorylated by c-Abl and promotes the apoptotic effect of PKCδ in glioma cells,” Biochemical and Biophysical Research Communications, vol. 352, no. 2, pp. 431–436, 2007.
[230]
R. Gomel, C. Xiang, S. Finniss et al., “The localization of protein kinase Cδ in different subcellular sites affects its proapoptotic and antiapoptotic functions and the activation of distinct downstream signaling pathways,” Molecular Cancer Research, vol. 5, no. 6, pp. 627–639, 2007.
[231]
H. Okhrimenko, W. Lu, C. Xiang et al., “Roles of tyrosine phosphorylation and cleavage of protein kinase Cδ in its protective effect against tumor necrosis factor-related apoptosis inducing ligand-induced apoptosis,” The Journal of Biological Chemistry, vol. 280, no. 25, pp. 23643–23652, 2005.
[232]
A. Zrachia, M. Dobroslav, M. Blass et al., “Infection of glioma cells with Sindbis virus induces selective activation and tyrosine phosphorylation of protein kinase C δ: implications for sindbis virus-induced apoptosis,” The Journal of Biological Chemistry, vol. 277, no. 26, pp. 23693–23701, 2002.
[233]
M. Blass, I. Kronfeld, G. Kazimirsky, P. M. Blumberg, and C. Brodie, “Tyrosine phosphorylation of protein kinase Cδ is essential for its apoptotic effect in response to etoposide,” Molecular and Cellular Biology, vol. 22, no. 1, pp. 182–195, 2002.
[234]
R. Mandil, E. Ashkenazi, M. Blass et al., “Protein kinase Cα and protein kinase Cδ play opposite roles in the proliferation and apoptosis of glioma cells,” Cancer Research, vol. 61, no. 11, pp. 4612–4619, 2001.
[235]
S. L. Lomonaco, S. Kahana, M. Blass et al., “Phosphorylation of protein kinase Cδ on distinct tyrosine residues induces sustained activation of Erk1/2 via down-regulation of MKP-1: role in the apoptotic effect of etoposide,” The Journal of Biological Chemistry, vol. 283, no. 25, pp. 17731–17739, 2008.
[236]
C. Peng, T. Tseng, J. Liu et al., “Penta-acetyl geniposide-induced C6 glioma cell apoptosis was associated with the activation of protein kinase C-delta,” Chemico-Biological Interactions, vol. 147, no. 3, pp. 287–296, 2004.
[237]
C. Peng, C. Huang, S. Hsu, and C. Wang, “Penta-acetyl geniposide induce apoptosis in C6 glioma cells by modulating the activation of neutral sphingomyelinase-induced p75 nerve growth factor receptor and protein kinase Cδ pathway,” Molecular Pharmacology, vol. 70, no. 3, pp. 997–1004, 2006.
[238]
S. Kim, J. Hwang, W. Lee, D. Y. Hwang, and K. Suk, “Role of protein kinase Cδ in paraquat-induced glial cell death,” Journal of Neuroscience Research, vol. 86, no. 9, pp. 2062–2070, 2008.
[239]
M. Kim, R. Kim, C. Yoon et al., “Importance of PKCδ signaling in fractionatedradiation-induced expansion of glioma-initiating cells and resistance to cancer treatment,” Journal of Cell Science, vol. 124, no. 18, pp. 3084–3094, 2011.
[240]
S. Amos, P. M. Martin, G. A. Polar, S. J. Parsons, and I. M. Hussaini, “Phorbol 12-myristate 13-acetate induces epidermal growth factor receptor transactivation via protein kinase Cδ/c-Src pathways in glioblastoma cells,” The Journal of Biological Chemistry, vol. 280, no. 9, pp. 7729–7738, 2005.
[241]
B. S. Paugh, S. W. Paugh, L. Bryan et al., “EGF regulates plasminogen activator inhibitor-1 (PAI-1) by a pathway involving c-Src, PKCδ, and sphingosine kinase 1 in glioblastoma cells,” The FASEB Journal, vol. 22, no. 2, pp. 455–465, 2008.
[242]
S. Sarkar and V. W. Yong, “Reduction of protein kinase C delta attenuates tenascin-C stimulated glioma invasion in three-dimensional matrix,” Carcinogenesis, vol. 31, no. 2, pp. 311–317, 2010.
[243]
A. Forastiere, W. Koch, A. Trotti, and D. Sidransky, “Head and neck cancer,” The New England Journal of Medicine, vol. 345, no. 26, pp. 1890–1900, 2001.
[244]
R. I. Haddad and D. M. Shin, “Recent advances in head and neck cancer,” The New England Journal of Medicine, vol. 359, no. 11, pp. 1096–1154, 2008.
[245]
E. E. W. Cohen, H. Zhu, M. W. Lingen et al., “A feed-forward loop involving protein kinase Cα and microRNAs regulates tumor cell cycle,” Cancer Research, vol. 69, no. 1, pp. 65–74, 2009.
[246]
F. Liu, Z. Zhao, P. Li et al., “NF-κB participates in chemokine receptor 7-mediated cell survival in metastatic squamous cell carcinoma of the head and neck,” Oncology Reports, vol. 25, no. 2, pp. 383–391, 2011.
[247]
Z. J. Zhao, P. Li, F. Y. Liu, L. Sun, and C. F. Sun, “PKCα take part in CCR7/NF-κB autocrine signaling loop in CCR7-positive squamous cell carcinoma of head and neck,” Molecular and Cellular Biochemistry, vol. 357, no. 1-2, pp. 181–187, 2011.
[248]
Q. Pan, L. W. Bao, T. N. Teknos, and S. D. Merajver, “Targeted disruption of protein kinase Cε reduces cell invasion and motility through inactivation of RhoA and RhoC GTPases in head and neck squamous cell carcinoma,” Cancer Research, vol. 66, no. 19, pp. 9379–9384, 2006.
[249]
J. Datta, A. Smith, J. C. Lang et al., “microRNA-107 functions as a candidate tumor-suppressor gene in head and neck squamous cell carcinoma by downregulation of protein kinase Ce{open},” Oncogene, vol. 31, no. 36, pp. 4045–4053, 2012.
[250]
E. E. W. Cohen, M. W. Lingen, B. Zhu et al., “Protein kinase Cζ mediates epidermal growth factor-induced growth of head and neck tumor cells by regulating mitogen-activated protein kinase,” Cancer Research, vol. 66, no. 12, pp. 6296–6303, 2006.
[251]
C. Valkova, C. Mertens, S. Weisheit, D. Imhof, and C. Liebmann, “Activation by tyrosine phosphorylation as a prerequisite for protein kinase Cζ to mediate epidermal growth factor receptor signaling to ERK,” Molecular Cancer Research, vol. 8, no. 5, pp. 783–797, 2010.
[252]
Z. Xie, Y. Jiang, E. Liao et al., “PIKE mediates EGFR proliferative signaling in squamous cell carcinoma cells,” Oncogene, vol. 31, no. 49, pp. 5090–5098, 2012.
[253]
K. C. Lai, C. J. Liu, K. W. Chang, and T. C. Lee, “Depleting IFIT2 mediates atypical PKC signaling to enhance the migration and metastatic activity of oral squamous cell carcinoma cells,” Oncogene, vol. 32, no. 32, pp. 3686–3697, 2013.
[254]
C.-C. Yu, S.-C. Lo, and T.-C. V. Wang, “Telomerase is regulated by protein kinase C-ζ in human nasopharyngeal cancer cells,” Biochemical Journal, vol. 355, no. 2, pp. 459–464, 2001.
[255]
M. J. Frederick, A. J. VanMeter, M. A. Gadhikar et al., “Phosphoproteomic analysis of signaling pathways in head and neck squamous cell carcinoma patient samples,” American Journal of Pathology, vol. 178, no. 2, pp. 548–571, 2011.
[256]
Y. Yang, J. Chu, M. Luo et al., “Amplification of PRKCI, located in 3q26, is associated with lymph node metastasis in esophageal squamous cell carcinoma,” Genes Chromosomes and Cancer, vol. 47, no. 2, pp. 127–136, 2008.
[257]
S. Liu, B. Wang, Y. Jiang et al., “Atypical protein kinase Cι (PKCι) promotes metastasis of esophageal squamous cell carcinoma by enhancing resistance to anoikis via PKCι-SKP2-AKT Pathway,” Molecular Cancer Research, vol. 9, no. 4, pp. 390–402, 2011.
[258]
B. S. Wang, Y. Yang, H. Yang, et al., “PKCι counteracts oxidative stress by regulating Hsc70 in an esophageal cancer cell line,” Cell Stress and Chaperones, vol. 18, no. 3, pp. 359–366, 2013.
[259]
P. Chu, N. C. Hsu, H. Tai et al., “High nuclear protein kinase Cθ expression may correlate with disease recurrence and poor survival in oral squamous cell carcinoma,” Human Pathology, vol. 43, no. 2, pp. 276–281, 2012.
[260]
P. Y. Chu, N. C. H. Hsu, S. H. Lin, M. F. How, and K. T. Yeh, “High nuclear protein kinase C βII expression is a marker of disease recurrence in oral squamous cell carcinoma,” Anticancer Research, vol. 32, no. 9, pp. 3987–3991, 2012.
[261]
L. Ducher, F. Croquet, S. Gil, J. Davy, J. Féger, and A. Bréhier, “Differential expression of five protein kinase C isoenzymes in FAO and HepG2 hepatoma cell lines compared with normal rat hepatocytes,” Biochemical and Biophysical Research Communications, vol. 217, no. 2, pp. 546–553, 1995.
[262]
S. Hsu, Y. Chou, S. Yin, and J. Liu, “Differential effects of phorbol ester on growth and protein kinase C isoenzyme regulation in human hepatoma Hep3B cells,” Biochemical Journal, vol. 333, part 1, pp. 57–64, 1998.
[263]
T. Ohigashi, C. S. Mallia, E. McGary et al., “Protein kinase C α protein expression is necessary for sustained erythropoietin production in human hepatocellular carcinoma (Hep3B) cells exposed to hypoxia,” Biochimica et Biophysica Acta, vol. 1450, no. 2, pp. 109–118, 1999.
[264]
H. Lu, F. Chou, K. Yeh, Y. Chang, N. C. Hsu, and J. Chang, “Expression of protein kinase C family in human hepatocellular carcinoma,” Pathology and Oncology Research, vol. 16, no. 3, pp. 385–391, 2010.
[265]
J. Wang, Q. Li, G. Du, J. Lu, and S. Zou, “Significance and expression of atypical protein kinase C-ι in human hepatocellular carcinoma,” Journal of Surgical Research, vol. 154, no. 1, pp. 143–149, 2009.
[266]
G. Du, J. Wang, J. Lu et al., “Expression of P-aPKC-ι, E-cadherin, and β-catenin related to invasion and metastasis in hepatocellular carcinoma,” Annals of Surgical Oncology, vol. 16, no. 6, pp. 1578–1586, 2009.
[267]
H. Lu, F. Chou, K. Yeh, Y. Chang, N. C. Hsu, and J. Chang, “Analysing the expression of protein kinase C eta in human hepatocellular carcinoma,” Pathology, vol. 41, no. 7, pp. 626–629, 2009.
[268]
T. Wu, Y. Hsieh, Y. Hsieh, and J. Liu, “Reduction of PKCα decreases cell proliferation, migration, and invasion of human malignant hepatocellular carcinoma,” Journal of Cellular Biochemistry, vol. 103, no. 1, pp. 9–20, 2008.
[269]
Y. Hsieh, T. Wu, C. Huang, Y. Hsieh, J. Hwang, and J. Liu, “p38 mitogen-activated protein kinase pathway is involved in protein kinase Cα-regulated invasion in human hepatocellular carcinoma cells,” Cancer Research, vol. 67, no. 9, pp. 4320–4327, 2007.
[270]
J. Tsai, Y. Hsieh, S. Kuo et al., “Alteration in the expression of protein kinase C isoforms in human hepatocellular carcinoma,” Cancer Letters, vol. 161, no. 2, pp. 171–175, 2000.
[271]
W. Wu, R. K. Tsai, C. H. Chang, S. Wang, J. Wu, and Y. Chang, “Reactive oxygen species mediated sustained activation of protein kinase C α and extracellular signal-regulated kinase for migration of human hepatoma cell Hepg2,” Molecular Cancer Research, vol. 4, no. 10, pp. 747–758, 2006.
[272]
W. S. Wu, “Protein kinase C α trigger Ras and Raf-independent MEK/ERK activation for TPA-induced growth inhibition of human hepatoma cell HepG2,” Cancer Letters, vol. 239, no. 1, pp. 27–35, 2006.
[273]
W. S. Wu and J. M. Huang, “Activation of protein kinase C alpha is required for TPA-triggered ERK (MAPK) signaling and growth inhibition of human hepatoma cell HepG2,” Journal of Biomedical Science, vol. 12, no. 2, pp. 289–296, 2005.
[274]
C. Chiang, Y. Huang, K. Leong et al., “PKCalpha mediated induction of miR-101 in human hepatoma HepG2 cells,” Journal of Biomedical Science, vol. 17, article 35, 2010.
[275]
H. F. Bunn, “Erythropoietin,” Cold Spring Harbor Perspectives in Medicine, vol. 3, no. 3, Article ID a011619, 2013.
[276]
C. T. Hu, C. C. Cheng, S. M. Pan, J. R. Wu, and W. S. Wu, “PKC mediates fluctuant ERK-paxillin signaling for hepatocyte growth factor-induced migration of hepatoma cell HepG2,” Cellular Signalling, vol. 25, no. 6, pp. 1457–1467, 2013.
[277]
T. Wu, Y. Hsieh, C. Wu, Y. Hsieh, C. Huang, and J. Liu, “Overexpression of protein kinase Cα mRNA in human hepatocellular carcinoma: a potential marker of disease prognosis,” Clinica Chimica Acta, vol. 382, no. 1-2, pp. 54–58, 2007.
[278]
C. Seeger and W. S. Mason, “Hepatitis B virus biology,” Microbiology and Molecular Biology Reviews, vol. 64, no. 1, pp. 51–68, 2000.
[279]
E. Hildt, B. Munz, G. Saher, K. Reifenberg, and P. H. Hofschneider, “The PreS2 activator MHB of hepatitis B virus activates c-raf-1/Erk2 signaling in transgenic mice,” The EMBO Journal, vol. 21, no. 4, pp. 525–535, 2002.
[280]
H. Liu, J. Xu, L. Zhou et al., “Hepatitis B virus large surface antigen promotes liver carcinogenesis by activating the Src/PI3K/Akt pathway,” Cancer Research, vol. 71, no. 24, pp. 7547–7557, 2011.
[281]
J. Hung, Y. Lu, Y. Wang et al., “FTY720 induces apoptosis in hepatocellular carcinoma cells through activation of protein kinase C δ signaling,” Cancer Research, vol. 68, no. 4, pp. 1204–1212, 2008.
[282]
C. Yoon, M. Kim, M. Park et al., “Claudin-1 acts through c-Abl-protein kinase Cδ (PKCδ) signaling and has a causal role in the acquisition of invasive capacity in human liver cells,” The Journal of Biological Chemistry, vol. 285, no. 1, pp. 226–233, 2010.
[283]
S. Takai, R. Matsushima-Nishiwaki, H. Tokuda et al., “Protein kinase C δ regulates the phosphorylation of heat shock protein 27 in human hepatocellular carcinoma,” Life Sciences, vol. 81, no. 7, pp. 585–591, 2007.
[284]
K. Guo, N. X. Kang, Y. Li et al., “Regulation of HSP27 on NF-κB pathway activation may be involved in metastatic hepatocellular carcinoma cells apoptosis,” BMC Cancer, vol. 9, article 100, 2009.
[285]
Y. Wang, D. Sun, and D. Liu, “Tumor suppression by RNA from C/EBPβ 3′UTR through the inhibition of protein kinase Cε activity,” PLoS ONE, vol. 6, no. 1, Article ID e16543, 2011.
[286]
K. Guo, Y. Li, X. Kang et al., “Role of PKCβ in hepatocellular carcinoma cells migration and invasion in vitro: a potential therapeutic target,” Clinical and Experimental Metastasis, vol. 26, no. 3, pp. 189–195, 2009.
[287]
R. S. Herbst, J. V. Heymach, and S. M. Lippman, “Molecular origins of cancer: lung cancer,” The New England Journal of Medicine, vol. 359, no. 13, pp. 1367–1380, 2008.
[288]
S. S. Singhal, S. Yadav, J. Singhal, K. Drake, Y. C. Awasthi, and S. Awasthi, “The role of PKCα and RLIP76 in transport-mediated doxorubicin-resistance in lung cancer,” FEBS Letters, vol. 579, no. 21, pp. 4635–4641, 2005.
[289]
W. Lang, H. Wang, L. Ding, and L. Xiao, “Cooperation between PKC-α and PKC-ε in the regulation of JNK activation in human lung cancer cells,” Cellular Signalling, vol. 16, no. 4, pp. 457–467, 2004.
[290]
M. Lahn, C. Su, S. Li et al., “Expression levels of protein kinase C-α in non-small-cell lung cancer,” Clinical Lung Cancer, vol. 6, no. 3, pp. 184–189, 2004.
[291]
A. K. O'Neill, L. L. Gallegos, V. Justilien et al., “Protein kinase Cα promotes cell migration through a PDZ-dependent interaction with its novel substrate discs large homolog 1 (DLG1),” The Journal of Biological Chemistry, vol. 286, no. 50, pp. 43559–43568, 2011.
[292]
C. A. M. la Porta, L. Tessitore, and R. Comolli, “Changes in protein kinase C α,δ and in nuclear β isoform expression in tumour and lung metastatic nodules induced by diethylnitrosamine in the rat,” Carcinogenesis, vol. 18, no. 4, pp. 715–719, 1997.
[293]
C. Wang, X. Wang, H. Liang, et al., “miR-203 inhibits cell proliferation and migration lung cancer cells by targeting PKCα,” PLoS ONE, vol. 8, no. 9, Article ID e73985, 2013.
[294]
D. Xiao, K. Wang, J. Zhou et al., “Inhibition of fibroblast growth factor 2-induced apoptosis involves survivin expression, protein kinase Cα activation and subcellular translocation of Smac in human small cell lung cancer cells,” Acta Biochimica et Biophysica Sinica, vol. 40, no. 4, pp. 297–303, 2008.
[295]
K. S. Hill, E. Erdogan, A. Khoor, et al., “Protein kinase Cα suppresses Kras-mediated lung tumor formation through activation of a p38 MAPK-TGFβ signaling axis,” Oncogene, 2013.
[296]
L. Paz-Ares, J. Douillard, P. Koralewski et al., “Phase III study of gemcitabine and cisplatin with or without aprinocarsen, a protein kinase C-alpha antisense oligonucleotide, in patients with advanced-stage non-small-cell lung cancer,” Journal of Clinical Oncology, vol. 24, no. 9, pp. 1428–1434, 2006.
[297]
P. Ritch, C. M. Rudin, J. D. Bitran et al., “Phase II study of PKC-α antisense oligonucleotide aprinocarsen in combination with gemcitabine and carboplatin in patients with advanced non-small cell lung cancer,” Lung Cancer, vol. 52, no. 2, pp. 173–180, 2006.
[298]
M. A. Villalona-Calero, P. Ritch, J. A. Figueroa et al., “A phase I/II study of LY900003, an antisense inhibitor of protein kinase C-α, in combination with cisplatin and gemcitabine in patients with advanced non-small cell lung cancer,” Clinical Cancer Research, vol. 10, no. 18, part 1, pp. 6086–6093, 2004.
[299]
K. Bae, H. Wang, G. Jiang, M. G. Chen, L. Lu, and L. Xiao, “Protein kinase Cε is overexpressed in primary human non-small cell lung cancers and functionally required for proliferation of non-small cell lung cancer cells in a p21/Cip1-dependent manner,” Cancer Research, vol. 67, no. 13, pp. 6053–6063, 2007.
[300]
M. C. Caino, C. Lopez-Haber, J. Kim, D. Mochly-Rosen, and M. G. Kazanietz, “Protein kinase C? is required for non-small cell lung carcinoma growth and regulates the expression of apoptotic genes,” Oncogene, vol. 31, no. 20, pp. 2593–2600, 2012.
[301]
M. Felber, J. Sonnemann, and J. F. Beck, “Inhibition of novel protein kinase Cε augments TRAIL-induced cell death in A549 lung cancer cells,” Pathology and Oncology Research, vol. 13, no. 4, pp. 295–301, 2007.
[302]
L. Ding, H. Wang, W. Lang, and L. Xiao, “Protein kinase C-ε promotes survival of lung cancer cells by suppressing apoptosis through dysregulation of the mitochondrial caspase pathway,” The Journal of Biological Chemistry, vol. 277, no. 38, pp. 35305–35313, 2002.
[303]
O. E. Pardo, C. Wellbrock, U. K. Khanzada et al., “FGF-2 protects small cell lung cancer cells from apoptosis through a complex involving PKCε, B-Raf and S6K2,” The EMBO Journal, vol. 25, no. 13, pp. 3078–3088, 2006.
[304]
M. C. Caino, C. Lopez-Haber, J. L. Kissil, and M. G. Kazanietz, “Non-small cell lung carcinoma cell motility, Rac activation and metastatic dissemination are mediated by protein kinase C epsilon,” PLoS ONE, vol. 7, no. 2, Article ID e31714, 2012.
[305]
S. Tuomi, A. Mai, J. Nevo et al., “PKCε regulation of an α5 integrin-ZO-1 complex controls lamellae formation in migrating cancer cells,” Science Signaling, vol. 2, no. 77, article ra32, 2009.
[306]
R. P. Regala, C. Weems, L. Jamieson et al., “Atypical protein kinase Cι is an oncogene in human non-small cell lung cancer,” Cancer Research, vol. 65, no. 19, pp. 8905–8911, 2005.
[307]
R. P. Regala, C. Weems, L. Jamieson, J. A. Copland, E. A. Thompson, and A. P. Fields, “Atypical protein kinase Cτ plays a critical role in human lung cancer cell growth and tumorigenicity,” The Journal of Biological Chemistry, vol. 280, no. 35, pp. 31109–31115, 2005.
[308]
S. Liu, B. Wang, Y. Jiang et al., “Atypical protein kinase Cι (PKCι) promotes metastasis of esophageal squamous cell carcinoma by enhancing resistance to anoikis via PKCι-SKP2-AKT pathway,” Molecular Cancer Research, vol. 9, no. 4, pp. 390–402, 2011.
[309]
L. Xu and X. Deng, “Protein kinase Cι promotes nicotine-induced migration and invasion of cancer cells via phosphorylation of μ- and m-calpains,” The Journal of Biological Chemistry, vol. 281, no. 7, pp. 4457–4466, 2006.
[310]
J. C. Shultz, N. Vu, M. D. Shultz, M. U. U. Mba, B. A. Shapiro, and C. E. Chalfant, “The proto-oncogene PKCι regulates the alternative splicing of Bcl-x pre-mRNA,” Molecular Cancer Research, vol. 10, no. 5, pp. 660–669, 2012.
[311]
R. P. Regala, E. A. Thompson, and A. P. Fields, “Atypical protein kinase Cι expression and aurothiomalate sensitivity in human lung cancer cells,” Cancer Research, vol. 68, no. 14, pp. 5888–5895, 2008.
[312]
V. Justilien, L. Jameison, C. J. Der, K. L. Rossman, and A. P. Fields, “Oncogenic activity of Ect2 is regulated through protein kinase Cι-mediated phosphorylation,” The Journal of Biological Chemistry, vol. 286, no. 10, pp. 8149–8157, 2011.
[313]
L. A. Frederick, J. A. Matthews, L. Jamieson et al., “Matrix metalloproteinase-10 is a critical effector of protein kinase Cι-Par6α-mediated lung cancer,” Oncogene, vol. 27, no. 35, pp. 4841–4853, 2008.
[314]
E. Erdogan, E. W. Klee, E. A. Thompson, and A. P. Fields, “Meta-analysis of oncogenic protein kinase Cι signaling in lung adenocarcinoma,” Clinical Cancer Research, vol. 15, no. 5, pp. 1527–1533, 2009.
[315]
D. Meng, M. Yuan, X. Li, et al., “Prognostic value of K-RAS mutations in patients with non-small cell lung cancer: a systematic review with meta-analysis,” Lung Cancer, vol. 81, no. 1, pp. 1–10, 2013.
[316]
N. Karachaliou, C. Mayo, C. Costa, et al., “KRAS mutations in lung cancer,” Clinical Lung Cancer, vol. 14, no. 3, pp. 205–214, 2013.
[317]
M. Stailings-Mann, L. Jamieson, R. P. Regala, C. Weems, N. R. Murray, and A. P. Fields, “A novel small-molecule inhibitor of protein kinase Cι blocks transformed growth of non-small-cell lung cancer cells,” Cancer Research, vol. 66, no. 3, pp. 1767–1774, 2006.
[318]
R. P. Regala, R. K. Davis, A. Kunz, A. Khoor, M. Leitges, and A. P. Fields, “Atypical protein kinase Cι is required for bronchioalveolar stem cell expansion and lung tumorigenesis,” Cancer Research, vol. 69, no. 19, pp. 7603–7611, 2009.
[319]
W. Guo, S. Wu, J. Liu, and B. Fang, “Identification of a small molecule with synthetic lethality for K-Ras and protein kinase C iota,” Cancer Research, vol. 68, no. 18, pp. 7403–7408, 2008.
[320]
E. Krasnitsky, Y. Baumfeld, J. Freedman et al., “PKCη is a novel prognostic marker in non-small cell lung cancer,” Anticancer Research, vol. 32, no. 4, pp. 1507–1513, 2012.
[321]
J. Sonnemann, V. Gekeler, K. Ahlbrecht et al., “Down-regulation of protein kinase Cη by antisense oligonucleotides sensitises A549 lung cancer cells to vincristine and paclitaxel,” Cancer Letters, vol. 209, no. 2, pp. 177–185, 2004.
[322]
Y. Liu, B. Wang, J. Wang et al., “Down-regulation of PKCζ expression inhibits chemotaxis signal transduction in human lung cancer cells,” Lung Cancer, vol. 63, no. 2, pp. 210–218, 2009.
[323]
J. M. Symonds, A. M. Ohm, C. J. Carter et al., “Protein kinase C δ is a downstream effector of oncogenic K-ras in lung tumors,” Cancer Research, vol. 71, no. 6, pp. 2087–2097, 2011.
[324]
A. S. Clark, K. A. West, P. M. Blumberg, and P. A. Dennis, “Altered protein kinase C (PKC) isoforms in non-small cell lung cancer cells: PKCδ promotes cellular survival and chemotherapeutic resistance,” Cancer Research, vol. 63, no. 4, pp. 780–786, 2003.
[325]
E. Kim, H. Lee, D. Lee et al., “Inhibition of heat shock protein 27-mediated resistance to DNA damaging agents by a novel PKCδ-V5 heptapeptide,” Cancer Research, vol. 67, no. 13, pp. 6333–6341, 2007.
[326]
L. Xu, L. Su, and X. Liu, “PKCδ regulates death receptor 5 expression induced by PS-341 through ATF4-ATF3/CHOP axis in human lung cancer cells,” Molecular Cancer Therapeutics, vol. 11, no. 10, pp. 2174–2182, 2012.
[327]
M. Nakagawa, J. L. Oliva, D. Kothapalli, A. Fournier, R. K. Assoian, and M. G. Kazanietz, “Phorbol ester-induced G1 phase arrest selectively mediated by protein kinase Cδ-dependent induction of p21,” The Journal of Biological Chemistry, vol. 280, no. 40, pp. 33926–33934, 2005.
[328]
S. S. Hecht, “Tobacco carcinogens, their biomarkers and tobacco-induced cancer,” Nature Reviews Cancer, vol. 3, no. 10, pp. 733–744, 2003.
[329]
H. Lemjabbar-Alaoui, S. S. Sidhu, A. Mengistab, M. Gallup, and C. Basbaum, “TACE/ADAM-17 phosphorylation by PKC-epsilon mediates premalignant changes in Tobacco smoke-exposed lung cells,” PLoS ONE, vol. 6, no. 3, Article ID e17489, 2011.
[330]
Y. M. Whang, U. Jo, J. S. Sung, et al., “Wnt5a is associated with cigarette smoke-related lung carcinogenesis via protein kinase C,” PLoS ONE, vol. 8, no. 1, Article ID e53012, 2013.
[331]
J. Shen, L. Xu, T. K. Owonikoko et al., “NNK promotes migration and invasion of lung cancer cells through activation of c-Src/PKCι/FAK loop,” Cancer Letters, vol. 318, no. 1, pp. 106–113, 2012.
[332]
R. Gopalakrishna, Z. Chen, and U. Gundimeda, “Tobacco smoke tumor promoters, catechol and hydroquinone, induce oxidative regulation of protein kinase C and influence invasion and metastasis of lung carcinoma cells,” Proceedings of the National Academy of Sciences of the United States of America, vol. 91, no. 25, pp. 12233–12237, 1994.
[333]
M. Oka and U. Kikkawa, “Protein kinase C in melanoma,” Cancer and Metastasis Reviews, vol. 24, no. 2, pp. 287–300, 2005.
[334]
M. F. Denning, “Specifying protein kinase C functions in melanoma,” Pigment Cell & Melanoma Research, vol. 25, no. 4, pp. 466–476, 2012.
[335]
M. M. Lahn and K. L. Sundell, “The role of protein kinase C-alpha (PKC-α) in melanoma,” Melanoma Research, vol. 14, no. 2, pp. 85–89, 2004.
[336]
S. D. Smith, M. Enge, W. Bao, et al., “Protein kinase Cα (PKCα) regulates p53 localization and melanoma cell survival downstream of integrin αv in three-dimensional collagen and in vivo,” The Journal of Biological Chemistry, vol. 287, no. 35, pp. 29336–29347, 2012.
[337]
A. J. Putnam, V. V. Schulz, E. M. Freiter, H. M. Bill, and C. K. Miranti, “Src, PKCα, and PKCδ are required for αvβ3 integrin-mediated metastatic melanoma invasion,” Cell Communication and Signalling, vol. 7, article 10, 2009.
[338]
H. R. Byers, S. J. S. Boissel, C. Tu, and H. Park, “RNAi-mediated knockdown of protein kinase C-alpha inhibits cell migration in MM-RU human metastatic melanoma cell line,” Melanoma Research, vol. 20, no. 3, pp. 171–178, 2010.
[339]
A. Vartanian, E. Stepanova, I. Grigorieva, E. Solomko, A. Baryshnikov, and M. Lichinitser, “VEGFR1 and PKCα signaling control melanoma vasculogenic mimicry in a VEGFR2 kinase-independent manner,” Melanoma Research, vol. 21, no. 2, pp. 91–98, 2011.
[340]
R. E. B. Seftor, A. R. Hess, E. A. Seftor, et al., “Tumor cell vasculogenic mimicry: from controversy to therapeutic promise,” The American Journal of Pathology, vol. 181, no. 4, pp. 1115–1125, 2012.
[341]
J. H. Kang, D. Asai, J. Oishi et al., “Role of plasma as activator and cofactor in phosphorylation catalyzed by protein kinase C,” Cell Biochemistry and Function, vol. 26, no. 1, pp. 70–75, 2008.
[342]
S. Gillespie, X. D. Zhang, and P. Hersey, “Variable expression of protein kinase Cε in human melanoma cells regulates sensitivity to TRAIL-induced apoptosis,” Molecular Cancer Therapeutics, vol. 4, no. 4, pp. 668–676, 2005.
[343]
N. M. Mhaidat, R. F. Thorne, X. D. Zhang, and P. Hersey, “Regulation of docetaxel-induced apoptosis of human melanoma cells by different isoforms of protein kinase C,” Molecular Cancer Research, vol. 5, no. 10, pp. 1073–1081, 2007.
[344]
E. Lau, H. Kluger, T. Varsano et al., “PKCε promotes oncogenic functions of ATF2 in the nucleus while blocking its apoptotic function at mitochondria,” Cell, vol. 148, no. 3, pp. 543–555, 2012.
[345]
M. Oka, U. Kikkawa, and C. Nishigori, “Protein kinase C-βII represses hepatocyte growth factor-induced invasion by preventing the association of adapter protein Gab1 and phosphatidylinositol 3-kinase in melanoma cells,” Journal of Investigative Dermatology, vol. 128, no. 1, pp. 188–195, 2008.
[346]
J. P. Voris, L. A. Sitailo, H. R. Rahn et al., “Functional alterations in protein kinase C beta II expression in melanoma,” Pigment Cell and Melanoma Research, vol. 23, no. 2, pp. 216–224, 2010.
[347]
C. A. M. la Porta and R. Comolli, “Overexpression of nPKCδ in BL6 murine melanoma cells enhances TGFβ1 release into the plasma of metastasized animals,” Melanoma Research, vol. 10, no. 6, pp. 527–534, 2000.
[348]
C. A. M. la Porta, A. di Dio, D. Porro, and R. Comolli, “Overexpression of novel protein kinase C δ in BL6 murine melanoma cells inhibits the proliferative capacity in vitro but enhances the metastatic potential in vivo,” Melanoma Research, vol. 10, no. 2, pp. 93–102, 2000.
[349]
E. Albi, C. A. M. la Porta, S. Cataldi, and M. V. Magni, “Nuclear sphingomyelin-synthase and protein kinase C δ in melanoma cells,” Archives of Biochemistry and Biophysics, vol. 438, no. 2, pp. 156–161, 2005.
[350]
N. M. Mhaidat, R. F. Thorne, D. Z. Xu, and P. Hersey, “Regulation of docetaxel-induced apoptosis of human melanoma cells by different isoforms of protein kinase C,” Molecular Cancer Research, vol. 5, no. 10, pp. 1073–1081, 2007.
[351]
S. T. Abrams, T. Lakum, K. Lin et al., “B-cell receptor signaling in chronic lymphocytic leukemia cells is regulated by overexpressed active protein kinase CβII,” Blood, vol. 109, no. 3, pp. 1193–1201, 2007.
[352]
N. N. Kabir, L. R?nnstrand, and J. U. Kazi, “Protein kinase C expression is deregulated in chronic lymphocytic leukemia,” Leukemia & Lymphoma, vol. 54, no. 10, pp. 2288–2290, 2013.
[353]
J. U. Kazi, N. N. Kabir, and L. R?nnstrand, “Protein kinase C, (PKC) as a drug target in chronic lymphocytic leukemia,” Medical Oncology, vol. 30, no. 4, article 757, 2013.
[354]
I. Espinosa, J. Briones, R. Bordes et al., “Membrane PKC-beta 2 protein expression predicts for poor response to chemotherapy and survival in patients with diffuse large B-cell lymphoma,” Annals of Hematology, vol. 85, no. 9, pp. 597–603, 2006.
[355]
R. Schaffel, J. C. Morais, I. Biasoli et al., “PKC-beta II expression has prognostic impact in nodal diffuse large B-cell lymphoma,” Modern Pathology, vol. 20, no. 3, pp. 326–330, 2007.
[356]
S. Riihij?rvi, S. Koivula, H. Nyman, K. Rydstr?m, M. Jerkeman, and S. Lepp?, “Prognostic impact of protein kinase C βII expression in R-CHOP-treated diffuse large B-cell lymphoma patients,” Modern Pathology, vol. 23, no. 5, pp. 686–693, 2010.
[357]
N. S. Saba and L. S. Levy, “Apoptotic induction in B-cell acute lymphoblastic leukemia cell lines treated with a protein kinase Cβ inhibitor,” Leukemia & Lymphoma, vol. 52, no. 5, pp. 877–886, 2011.
[358]
S. T. Abrams, B. R. B. Brown, M. Zuzel, and J. R. Slupsky, “Vascular endothelial growth factor stimulates protein kinase C βII expression in chronic lymphocytic leukemia cells,” Blood, vol. 115, no. 22, pp. 4447–4454, 2010.
[359]
M. Barragán, M. de Frias, D. Iglesias-Serret et al., “Regulation of Akt/PKB by phosphatidylinositol 3-kinase-dependent and -independent pathways in B-cell chronic lymphocytic leukemia cells: role of protein kinase Cβ,” Journal of Leukocyte Biology, vol. 80, no. 6, pp. 1473–1479, 2006.
[360]
C. Holler, J. D. Pi?ón, U. Denk et al., “PKCβ is essential for the development of chronic lymphocytic leukemia in the TCL1 transgenic mouse model: validation of PKCβ as a therapeutic target in chronic lymphocytic leukemia,” Blood, vol. 113, no. 12, pp. 2791–2794, 2009.
[361]
A. Patke, I. Mecklenbr?uker, H. Erdjument-Bromage, P. Tempst, and A. Tarakhovsky, “BAFF controls B cell metabolic fitness through a PKCβ- and Akt-dependent mechanism,” Journal of Experimental Medicine, vol. 203, no. 11, pp. 2551–2562, 2006.
[362]
C. M. zum Büschenfelde, M. Wagner, G. Lutzny et al., “Recruitment of PKC-βII to lipid rafts mediates apoptosis-resistance in chronic lymphocytic leukemia expressing ZAP-70,” Leukemia, vol. 24, no. 1, pp. 141–152, 2010.
[363]
G. Lutzny, T. Kocher, M. Schmidt-Supprian, et al., “Protein kinase C-β-dependent activation of NF-κB in stromal cells is indispensable for the survival of chronic lymphocytic leukemia B cells in vivo,” Cancer Cell, vol. 23, no. 1, pp. 77–92, 2013.
[364]
I. Ringshausen, F. Schneller, C. Bogner et al., “Constitutively activated phosphatidylinositol-3 kinase (PI-3K) is involved in the defect of apoptosis in B-CLL: association with protein kinase Cδ,” Blood, vol. 100, no. 10, pp. 3741–3748, 2002.
[365]
A. D. Baudot, P. Y. Jeandel, X. Mouska et al., “The tyrosine kinase Syk regulates the survival of chronic lymphocytic leukemia B cells through PKC and proteasome-dependent regulation of Mcl-1 expression,” Oncogene, vol. 28, no. 37, pp. 3261–3273, 2009.
[366]
R. Hubmann, M. Düchler, S. Schnabl et al., “NOTCH2 links protein kinase C delta to the expression of CD23 in chronic lymphocytic leukaemia (CLL) cells: research paper,” British Journal of Haematology, vol. 148, no. 6, pp. 868–878, 2010.
[367]
T. Jiffar, S. Kurinna, G. Suck et al., “PKC α mediates chemoresistance in acute lymphoblastic leukemia through effects on Bcl2 phosphorylation,” Leukemia, vol. 18, no. 3, pp. 505–512, 2004.
[368]
N. R. Murray, G. P. Baumgardner, D. J. Burns, and A. P. Fields, “Protein kinase C isotypes in human erythroleukemia (K562) cell proliferation and differentiation. Evidence that β(II) protein kinase C is required for proliferation,” The Journal of Biological Chemistry, vol. 268, no. 21, pp. 15847–15853, 1993.
[369]
M. Kaneki, S. Kharbanda, P. Pandey et al., “Functional role for protein kinase Cβ as a regulator of stress- activated protein kinase activation and monocytic differentiation of myeloid leukemia cells,” Molecular and Cellular Biology, vol. 19, no. 1, pp. 461–470, 1999.
[370]
P. P. Ruvolo, L. Zhou, J. C. Watt et al., “Targeting PKC-mediated signal transduction pathways using enzastaurin to promote apoptosis in acute myeloid leukemia-derived cell lines and blast cells,” Journal of Cellular Biochemistry, vol. 112, no. 6, pp. 1696–1707, 2011.
[371]
X. Li, Y. Li, Y. Zhang et al., “A novel bisindolymaleimide derivative (WK234) inhibits proliferation and induces apoptosis through the protein kinase Cβ pathway, in chronic myelogenous leukemia K562 cells,” Leukemia & Lymphoma, vol. 52, no. 7, pp. 1312–1320, 2011.
[372]
F. Pellicano, M. Copland, H. G. Jorgensen, J. Mountford, B. Leber, and T. L. Holyoake, “BMS-214662 induces mitochondrial apoptosis in chronic myeloid leukemia (CML) stem/progenitor cells, including CD34+38? cells, through activation of protein kinase Cβ,” Blood, vol. 114, no. 19, pp. 4186–4196, 2009.
[373]
A. Cataldi, M. Rapino, L. Centurione et al., “NF-κB activation plays an antiapoptotic role in human leukemic K562 cells exposed to ionizing radiation,” Journal of Cellular Biochemistry, vol. 89, no. 5, pp. 956–963, 2003.
[374]
S. Kaur, S. Parmar, J. Smith et al., “Role of protein kinase C-δ (PKC-δ) in the generation of the effects of IFN-α in chronic myelogenous leukemia cells,” Experimental Hematology, vol. 33, no. 5, pp. 550–557, 2005.
[375]
V. di Giacomo, M. Rapino, S. Miscia, C. di Giulio, and A. Cataldi, “Dual role of HIF-1α in delivering a survival or death signal in hypoxia exposed human K562 erythroleukemia cells,” Cell Biology International, vol. 33, no. 1, pp. 49–56, 2009.
[376]
Z. Li, K. Shi, L. Guan, Q. Jiang, Y. Yang, and C. Xu, “Downregulation of protein kinase Cα was involved in selenite-induced apoptosis of NB4 cells,” Oncology Research, vol. 19, no. 2, pp. 77–83, 2010.
[377]
S. Kurinna, M. Konopleva, S. L. Palla et al., “Bcl2 phosphorylation and active PKC α are associated with poor survival in AML,” Leukemia, vol. 20, no. 7, pp. 1316–1319, 2006.
[378]
S. M. Kornblau, H. T. Vu, P. Ruvolo et al., “BAX and PKCα modulate the prognostic impact of BCL2 expression in acute myelogenous leukemia,” Clinical Cancer Research, vol. 6, no. 4, pp. 1401–1409, 2000.
[379]
P. P. Ruvolo, X. Deng, B. K. Carr, and W. S. May, “A functional role for mitochondrial protein kinase Cα in Bcl2 phosphorylation and suppression of apoptosis,” The Journal of Biological Chemistry, vol. 273, no. 39, pp. 25436–25442, 1998.
[380]
F. Gao, Y. Wu, M. Zhao, C. Liu, L. Wang, and G. Chen, “Protein kinase C-δ mediates down-regulation of heterogeneous nuclear ribonucleoprotein K protein: involvement in apoptosis induction,” Experimental Cell Research, vol. 315, no. 19, pp. 3250–3258, 2009.
[381]
B. Ozpolat, U. Akar, I. Tekedereli, et al., “PKCδ regulates translation initiation through PKR and eIF2α in response to retinoic acid in acute myeloid leukemia cells,” Leukemia Research and Treatment, vol. 2012, Article ID 482905, 17 pages, 2012.
[382]
S. Wang, Y. Zheng, Y. Yu et al., “Phosphorylation of β-actin by protein kinase C-delta in camptothecin analog-induced leukemic cell apoptosis,” Acta Pharmacologica Sinica, vol. 29, no. 1, pp. 135–142, 2008.
[383]
H. Yan, Y.-C. Wang, D. Li et al., “Arsenic trioxide and proteasome inhibitor bortezomib synergistically induce apoptosis in leukemic cells: the role of protein kinase Cδ,” Leukemia, vol. 21, no. 7, pp. 1488–1495, 2007.
[384]
M. Song, S. Gao, K. Du et al., “Nanomolar concentration of NSC606985, a camptothecin analog, induces leukemic-cell apoptosis through protein kinase Cδ-dependent mechanisms,” Blood, vol. 105, no. 9, pp. 3714–3721, 2005.
[385]
B. Jang, K. Lim, J. Paik et al., “Tetrandrine-induced apoptosis is mediated by activation of caspases and PKC-δ in U937 cells,” Biochemical Pharmacology, vol. 67, no. 10, pp. 1819–1829, 2004.
[386]
H. Zhang, Y. Yang, K. Zhang et al., “Wogonin induced differentiation and G1 phase arrest of human U-937 leukemia cells via PKCδ phosphorylation,” European Journal of Pharmacology, vol. 591, no. 1–3, pp. 7–12, 2008.
[387]
A. Sassano, J. K. Altman, L. I. Gordon, and L. C. Platanias, “Statin-dependent activation of protein kinase Cδ in acute promyelocytic leukemia cells and induction of leukemic cell differentiation,” Leukemia & Lymphoma, vol. 53, no. 9, pp. 1779–1784, 2012.
[388]
A. Uruno, N. Noguchi, K. Matsuda et al., “All-trans retinoic acid and a novel synthetic retinoid tamibarotene (Am80) differentially regulate CD38 expression in human leukemia HL-60 cells: possible involvement of protein kinase C-δ,” Journal of Leukocyte Biology, vol. 90, no. 2, pp. 235–247, 2011.
[389]
K. Ohtani, H. Sakamoto, A. Kikuchi et al., “Follicle-stimulating hormone promotes the growth of human epithelial ovarian cancer cells through the protein kinase C-mediated system,” Cancer Letters, vol. 166, no. 2, pp. 207–213, 2001.
[390]
C. Mahanivong, H. M. Chen, S. W. Yee, Z. K. Pan, Z. Dong, and S. Huang, “Protein kinase Cα-CARMA3 signaling axis links Ras to NF-κB for lysophosphatidic acid-induced urokinase plasminogen activator expression in ovarian cancer cells,” Oncogene, vol. 27, no. 9, pp. 1273–1280, 2008.
[391]
W. Weichert, V. Gekeler, C. Denkert, M. Dietel, and S. Hauptmann, “Protein kinase C isoform expression in ovarian carcinoma correlates with indicator of poor prognosis,” International Journal of Oncology, vol. 23, no. 3, pp. 633–639, 2003.
[392]
N. Zhang, H. Zhang, L. Xia et al., “NSC606985 induces apoptosis, exerts synergistic effects with cisplatin, and inhibits hypoxia-stabilized HIF-1α protein in human ovarian cancer cells,” Cancer Letters, vol. 278, no. 2, pp. 139–144, 2009.
[393]
A. M. Eder, X. Sui, D. G. Rosen et al., “Atypical PKCι contributes to poor prognosis through loss of apical-basal polarity and cyclin E overexpression in ovarian cancer,” Proceedings of the National Academy of Sciences of the United States of America, vol. 102, no. 35, pp. 12519–12524, 2005.
[394]
I. Nazarenko, M. Jenny, J. Keil et al., “Atypical protein kinase C ζ exhibits a proapoptotic function in ovarian cancer,” Molecular Cancer Research, vol. 8, no. 6, pp. 919–934, 2010.
[395]
I. Mertens-Walker, C. Bolitho, R. C. Baxter, and D. J. Marsh, “Gonadotropin-induced ovarian cancer cell migration and proliferation require extracellular signal-regulated kinase 1/2 activation regulated by calcium and protein kinase Cδ,” Endocrine-Related Cancer, vol. 17, no. 2, pp. 335–349, 2010.
[396]
J. Yang and R. A. Weinberg, “Epithelial-mesenchymal transition: at the crossroads of development and tumor metastasis,” Developmental Cell, vol. 14, no. 6, pp. 818–829, 2008.
[397]
D. Kyuno, T. Kojima, T. Ito et al., “Protein kinase Cα inhibitor enhances the sensitivity of human pancreatic cancer HPAC cells to Clostridium perfringens enterotoxin via claudin-4,” Cell and Tissue Research, vol. 346, no. 3, pp. 369–381, 2011.
[398]
D. Kyuno, T. Kojima, H. Yamaguchi, et al., “Protein kinase Cα inhibitor protects against downregulation of claudin-1 during epithelial-mesenchymal transition of pancreatic cancer,” Carcinogenesis, vol. 34, no. 6, pp. 1232–1243, 2013.
[399]
Y. Chen, G. Yu, D. Yu, and M. Zhu, “PKCα-induced drug resistance in pancreatic cancer cells is associated with transforming growth factor-β1,” Journal of Experimental and Clinical Cancer Research, vol. 29, no. 1, article 104, 2010.
[400]
J. Y. C. Chow, H. Dong, K. T. Quach, P. N. van Nguyen, K. Chen, and J. M. Carethers, “TGF-β mediates PTEN suppression and cell motility through calcium-dependent PKC-α activation in pancreatic cancer cells,” American Journal of Physiology—Gastrointestinal and Liver Physiology, vol. 294, no. 4, pp. G899–G905, 2008.
[401]
M. G. Franz, J. G. Norman, P. J. Fabri, and W. R. Gower Jr., “Differentiation of pancreatic ductal carcinoma cells associated with selective expression of protein kinase C isoforms,” Annals of Surgical Oncology, vol. 3, no. 6, pp. 564–569, 1996.
[402]
S. Rosewicz, F. Brembeck, A. Kaiser, Z. V. Marschall, and E. Riecken, “Differential growth regulation by all-trans retinoic acid is determined by protein kinase C α in human pancreatic carcinoma cells,” Endocrinology, vol. 137, no. 8, pp. 3340–3347, 1996.
[403]
D. W. Denham, M. G. Franz, W. Denham et al., “Directed antisense therapy confirms the role of protein kinase C-α in the tumorigenicity of pancreatic cancer,” Surgery, vol. 124, no. 2, pp. 218–224, 1998.
[404]
K. M. Detjen, F. H. Brembeck, M. Welzel et al., “Activation of protein kinase Cα inhibits growth of pancreatic cancer cells via p2 -mediated G1 arrest,” Journal of Cell Science, vol. 113, no. 17, pp. 3025–3035, 2000.
[405]
A. M. Butler, M. L. S. Buzhardt, S. Li, K. E. Smith, A. P. Fields, and N. R. Murray, “Protein kinase C zeta regulates human pancreatic cancer cell transformed growth and invasion through a STAT3-dependent mechanism,” PLoS ONE, vol. 8, no. 8, Article ID e72061, 2013.
[406]
C. Laudanna, C. Sorio, C. Tecchio et al., “Motility analysis of pancreatic adenocarcinoma cells reveals a role for the atypical ζ isoform of protein kinase C in cancer cell movement,” Laboratory Investigation, vol. 83, no. 8, pp. 1155–1163, 2003.
[407]
M. D. Peruta, C. Giagulli, C. Laudanna, A. Scarpa, and C. Sorio, “RHOA and PRKCZ control different aspects of cell motility in pancreatic cancer metastatic clones,” Molecular Cancer, vol. 9, article 61, 2010.
[408]
M. Neid, K. Datta, S. Stephan et al., “Role of insulin receptor substrates and protein kinase C-ζ in vascular permeability factor/vascular endothelial growth factor expression in pancreatic cancer cells,” The Journal of Biological Chemistry, vol. 279, no. 6, pp. 3941–3948, 2004.
[409]
M. L. Scotti, W. R. Bamlet, T. C. Smyrk, A. P. Fields, and N. R. Murray, “Protein kinase Cι is required for pancreatic cancer cell transformed growth and tumorigenesis,” Cancer Research, vol. 70, no. 5, pp. 2064–2074, 2010.
[410]
S. Kato, K. Akimoto, Y. Nagashima, et al., “aPKCλ/ι is a beneficial prognostic marker for pancreatic neoplasms,” Pancreatology, vol. 13, no. 4, pp. 360–368, 2013.
[411]
M. L. Scotti, K. E. Smith, A. M. Butler et al., “Protein kinase C iota regulates pancreatic acinar-to-ductal metaplasia,” PLoS ONE, vol. 7, no. 2, Article ID e30509, 2012.
[412]
S. M. Cirigliano, L. V. Mauro, V. C. Grossoni, et al., “Modulation of pancreatic tumor potential by overexpression of protein kinase C β1,” Pancreas, vol. 42, no. 7, pp. 1060–1069, 2013.
[413]
A. C. Spalding, R. Watson, M. E. Davis, A. C. Kim, T. S. Lawrence, and E. Ben-Josef, “Inhibition of protein kinase Cβ by enzastaurin enhances radiation cytotoxicity in pancreatic cancer,” Clinical Cancer Research, vol. 13, no. 22, pp. 6827–6833, 2007.
[414]
A. C. Spalding, B. D. Zeitlin, K. Wilder-Romans, et al., “Enzastaurin, an inhibitor of PKCβ, enhances antiangiogenic effects and cytotoxicity of radiation against endothelial cells,” Translational Oncology, vol. 1, no. 4, pp. 195–201, 2008.
[415]
D. Molè, T. Gagliano, E. Gentilin et al., “Targeting protein kinase C by Enzastaurin restrains proliferation and secretion in human pancreatic endocrine tumors,” Endocrine-Related Cancer, vol. 18, no. 4, pp. 439–450, 2011.
[416]
L. V. Mauro, V. C. Grossoni, A. J. Urtreger et al., “PKC delta (PKCδ) promotes tumoral progression of human ductal pancreatic cancer,” Pancreas, vol. 39, no. 1, pp. e31–e41, 2010.
[417]
B. Ozpolat, U. Akar, K. Mehta, and G. Lopei-Berestein, “PKCδ and tissue transglutaminase are novel inhibitors of autophagy in pancreatic cancer cells,” Autophagy, vol. 3, no. 5, pp. 480–483, 2007.
[418]
P. Cornford, J. Evans, A. Dodson et al., “Protein kinase C isoenzyme patterns characteristically modulated in early prostate cancer,” American Journal of Pathology, vol. 154, no. 1, pp. 137–144, 1999.
[419]
J. Villar, M. I. Arenas, C. M. MacCarthy, M. J. Blánquez, O. M. Tirado, and V. Notario, “PCPH/ENTPD5 expression enhances the invasiveness of human prostate cancer cells by a protein kinase Cδ-dependent mechanism,” Cancer Research, vol. 67, no. 22, pp. 10859–10868, 2007.
[420]
C. Castilla, D. Chinchón, R. Medina, F. J. Torrubia, M. A. Japón, and C. Sáez, “PTPL1 and PKCδ contribute to proapoptotic signalling in prostate cancer cells,” Cell Death and Disease, vol. 4, article e576, 2013.
[421]
R. Koren, D. Ben Meir, L. Langzam et al., “Expression of protein kinase C isoenzymes in benign hyperplasia and carcinoma of prostate,” Oncology Reports, vol. 11, no. 2, pp. 321–326, 2004.
[422]
C. T. Powell, N. J. Brittis, D. Stec, H. Hug, W. D. W. Heston, and W. R. Fair, “Persistent membrane translocation of protein kinase C α during 12-O-tetradecanoylphorbol-13-acetate-induced apoptosis of LNCaP human prostate cancer cells,” Cell Growth & Differentiation, vol. 7, no. 4, pp. 419–428, 1996.
[423]
C. T. Powell and L. Yin, “Overexpression of PKCε sensitizes LNCaP human prostate cancer cells to induction of apoptosis by bryostatin 1,” International Journal of Cancer, vol. 118, no. 6, pp. 1572–1576, 2006.
[424]
B. Liu, R. J. Maher, J. P. de Jonckheere et al., “12(S)-HETE increases the motility of prostate tumor cells through selective activation of PKC(α),” Advances in Experimental Medicine and Biology, vol. 400B, pp. 707–718, 1997.
[425]
M. L. G. Lamm, D. D. Long, S. M. Goodwin, and C. Lee, “Transforming growth factor-β1 inhibits membrane association of protein kinase Cα in a human prostate cancer cell line, PC3,” Endocrinology, vol. 138, no. 11, pp. 4657–4664, 1997.
[426]
J. R. Stewart and C. A. O'Brian, “Protein kinase C-α mediates epidermal growth factor receptor transactivation in human prostate cancer cells,” Molecular Cancer Therapeutics, vol. 4, no. 5, pp. 726–732, 2005.
[427]
O. M. Fischer, S. Hart, A. Gschwind, and A. Ullrich, “EGFR signal transactivation in cancer cells,” Biochemical Society Transactions, vol. 31, no. 6, pp. 1203–1208, 2003.
[428]
S. Jain, G. Chakraborty, and G. C. Kundu, “The crucial role of cyclooxygenase-2 in osteopontin-induced protein kinase C α/c-Src/IκB kinase α/β-dependent prostate tumor progression and angiogenesis,” Cancer Research, vol. 66, no. 13, pp. 6638–6648, 2006.
[429]
J. Villar, H. S. Quadri, I. Song, Y. Tomita, O. M. Tirado, and V. Notario, “PCPH/ENTPD5 expression confers to prostate cancer cells resistance against cisplatin-induced apoptosis through protein kinase Cα-mediated Bcl-2 stabilization,” Cancer Research, vol. 69, no. 1, pp. 102–110, 2009.
[430]
A. Shih, S. Zhang, H. J. Cao et al., “Inhibitory effect of epidermal growth factor on resveratrol-induced apoptosis in prostate cancer cells is mediated by protien kinase C-α,” Molecular Cancer Therapeutics, vol. 3, no. 11, pp. 1355–1363, 2004.
[431]
J. Truman, S. A. Rotenberg, J. Kang et al., “PKCα activation downregulates ATM and radio-sensitizes androgen-sensitive human prostate cancer cells in vitro and in vivo,” Cancer Biology and Therapy, vol. 8, no. 1, pp. 54–63, 2009.
[432]
T. Kuo, W. Huang, and J. Guh, “WJ9708012 exerts anticancer activity through PKC-α related crosstalk of mitochondrial and endoplasmic reticulum stresses in human hormone-refractory prostate cancer cells,” Acta Pharmacologica Sinica, vol. 32, no. 1, pp. 89–98, 2011.
[433]
J. E. Gschwend, W. R. Fair, and C. T. Powell, “Bryostatin 1 induces prolonged activation of extracellular regulated protein kinases in and apoptosis of LNCaP human prostate cancer cells overexpressing protein kinase Cα,” Molecular Pharmacology, vol. 57, no. 6, pp. 1224–1234, 2000.
[434]
S. Rakoff-Nahoum and R. Medzhitov, “Toll-like receptors and cancer,” Nature Reviews Cancer, vol. 9, no. 1, pp. 57–63, 2009.
[435]
L. A. Rindour, R. Y. Cheng, C. H. Switzer, et al., “Molecular pathways: toll-like receptors in the tumor microenvironment-poor prognosis or new therapeutic opportunity,” Clinical Cancer Research, vol. 19, no. 6, pp. 1340–1346, 2013.
[436]
A. Paone, D. Starace, R. Galli et al., “Toll-like receptor 3 triggers apoptosis of human prostate cancer cells through a PKC-α-dependent mechanism,” Carcinogenesis, vol. 29, no. 7, pp. 1334–1342, 2008.
[437]
A. W. Tolcher, L. Reyno, P. M. Venner et al., “A randomized Phase II and pharmacokinetic study of the antisense oligonucleotides ISIS 3521 and ISIS 5132 in patients with hormone-refractory prostate cancer,” Clinical Cancer Research, vol. 8, no. 8, pp. 2530–2535, 2002.
[438]
B. B. Hafeez, W. Zhong, J. Weichert, N. E. Dreckschmidt, M. S. Jamal, and A. K. Verma, “Genetic ablation of PKC epsilon inhibits prostate cancer development and metastasis in transgenic mouse model of prostate adenocarcinoma,” Cancer Research, vol. 71, no. 6, pp. 2318–2327, 2011.
[439]
J. Chen, F. Deng, S. V. Singh, and Q. J. Wang, “Protein kinase D3 (PKD3) contributes to prostate cancer cell growth and survival through a PKCε/PKD3 pathway downstream of Akt and ERK 1/2,” Cancer Research, vol. 68, no. 10, pp. 3844–3853, 2008.
[440]
D. Wu, C. U. Thakore, G. G. Wescott, J. A. McCubrey, and D. M. Terrian, “Integrin signaling links protein kinase Cε to the protein kinase B/Akt survival pathway in recurrent prostate cancer cells,” Oncogene, vol. 23, no. 53, pp. 8659–8672, 2004.
[441]
D. Wu, T. L. Foreman, C. W. Gregory et al., “Protein kinase Cε has the potential to advance the recurrence of human prostate cancer,” Cancer Research, vol. 62, no. 8, pp. 2423–2429, 2002.
[442]
D. Wu and D. M. Terrian, “Regulation of caveolin-1 expression and secretion by a protein kinase Cε signaling pathway in human prostate cancer cells,” The Journal of Biological Chemistry, vol. 277, no. 43, pp. 40449–40455, 2002.
[443]
M. H. Aziz, H. T. Manoharan, D. R. Church et al., “Protein kinase Cε interacts with signal transducers and activators of transcription 3 (Stat3), phosphorylates Stat3Ser727, and regulates its constitutive activation in prostate cancer,” Cancer Research, vol. 67, no. 18, pp. 8828–8838, 2007.
[444]
J. Meshki, M. C. Caino, V. A. von Burstin, E. Griner, and M. G. Kazanietz, “Regulation of prostate cancer cell survival by protein kinase Cε involves bad phosphorylation and modulation of the TNFα/JNK pathway,” The Journal of Biological Chemistry, vol. 285, no. 34, pp. 26033–26040, 2010.
[445]
R. Garg, J. Blando, C. J. Perez, H. B. Wang, F. J. Benavides, and M. G. Kazaniets, “Activation of nuclear factor κB (NF-κB) in prostate cancer is mediated by protein kinase C ? (PKC?),” The Journal of Biological Chemistry, vol. 287, no. 44, pp. 37570–37582, 2012.
[446]
M. A. McJilton, C. van Sikes, G. G. Wescott et al., “Protein kinase Cε interacts with Bax and promotes survival of human prostate cancer cells,” Oncogene, vol. 22, no. 39, pp. 7958–7968, 2003.
[447]
E. Flescher and R. Rotem, “Protein kinase C ε mediates the induction of P-glycoprotein in LNCaP prostate carcinoma cells,” Cellular Signalling, vol. 14, no. 1, pp. 37–43, 2002.
[448]
S. Sarveswaran and J. Ghosh, “Wedelolactone, a medicinal plant-derived coumestan, induces caspase-dependent apoptosis in prostate cancer cells via downregulation of PKCε without inhibiting Akt,” International Journal of Oncology, vol. 41, no. 6, pp. 2191–2199, 2012.
[449]
U. Gundimeda, J. E. Schiffman, D. Chhabra, J. Wong, A. Wu, and R. Gopalakrishna, “Locally generated methylseleninic acid induces specific inactivation of protein kinase C isoenzymes: relevance to selenium-induced apoptosis in prostate cancer cells,” The Journal of Biological Chemistry, vol. 283, no. 50, pp. 34519–34531, 2008.
[450]
S. Sarveswaran, V. Thamilselvan, C. Brodie, and J. Ghosh, “Inhibition of 5-lipoxygenase triggers apoptosis in prostate cancer cells via down-regulation of protein kinase C-epsilon,” Biochimica et Biophysica Acta, vol. 1813, no. 12, pp. 2108–2117, 2011.
[451]
B. B. Hafeez, W. Zhong, A. Mustafa, J. W. Fischer, O. Witkowsky, and A. K. Verma, “Plumbagin inhibits prostate cancer development in TRAMP mice via targeting PKCε, Stat3 and neuroendocrine markers,” Carcinogenesis, vol. 33, no. 12, pp. 2586–2592, 2012.
[452]
J. Y. Kim, T. Valencia, S. Abu-Baker, et al., “c-Myc phosphorylation by PKCζ represses prostate tumorigenesis,” Proceedings of the National Academy of Sciences of the United States of America, vol. 110, no. 16, pp. 6418–6423, 2013.
[453]
A. M. Sánchez, S. Malagarie-Cazenave, N. Olea, D. Vara, C. Cuevas, and I. Díaz-Laviada, “Spisulosine (ES-285) induces prostate tumor PC-3 and LNCaP cell death by de novo synthesis of ceramide and PKCζ activation,” European Journal of Pharmacology, vol. 584, no. 2-3, pp. 237–245, 2008.
[454]
S. Yao, A. Bee, D. Brewer et al., “Prkc-ζ expression promotes the aggressive phenotype of human prostate cancer cells and is a novel target for therapeutic intervention,” Genes & Cancer, vol. 1, no. 5, pp. 444–464, 2010.
[455]
C. T. Powell, J. E. Gschwend, W. R. Fair, N. J. Brittis, D. Stec, and R. Huryk, “Overexpression of protein kinase C-zeta (PKC-ζ) inhibits invasive and metastatic abilities of Dunning R-3327 MAT-LyLu rat prostate cancer cells,” Cancer Research, vol. 56, no. 18, pp. 4137–4141, 1996.
[456]
T. Inoue, T. Yoshida, Y. Shimizu et al., “Requirement of androgen-dependent activation of protein kinase Cζ for androgen-dependent cell proliferation in LNCaP cells and its roles in transition to androgen-independent cells,” Molecular Endocrinology, vol. 20, no. 12, pp. 3053–3069, 2006.
[457]
J. Sonnemann, V. Gekeler, A. Sagrauske, C. Müller, H. Hofmann, and J. F. Beck, “Down-regulation of protein kinase Cη potentiates the cytotoxic effects of exogenous tumor necrosis factor-related apoptosis-inducing ligand in PC-3 prostate cancer cells,” Molecular Cancer Therapeutics, vol. 3, no. 7, pp. 773–781, 2004.
[458]
J. Kim, Y. Choi, A. Vallentin et al., “Centrosomal PKCβII and pericentrin are critical for human prostate cancer growth and angiogenesis,” Cancer Research, vol. 68, no. 16, pp. 6831–6839, 2008.
[459]
P. Lu, C. Yu, P. Chiang et al., “Paclitaxel induces apoptosis through activation of nuclear protein kinase C-δ and subsequent activation of Golgi associated Cdk1 in human hormone refractory prostate cancer,” The Journal of Urology, vol. 186, no. 6, pp. 2434–2441, 2011.
[460]
H. Wang, L. Xiao, and M. G. Kazanietz, “p23/Tmp21 associates with protein kinase Cδ (PKCδ) and modulates its apoptotic function,” The Journal of Biological Chemistry, vol. 286, no. 18, pp. 15821–15831, 2011.
[461]
M. V. Gavrielides, A. M. Gonzalez-Guerrico, N. A. Riobo, and M. G. Kazanietz, “Androgens regulate protein kinase Cδ transcription and modulate its apoptotic function in prostate cancer cells,” Cancer Research, vol. 66, no. 24, pp. 11792–11801, 2006.
[462]
A. M. Gonzalez-Guerrico and M. G. Kazanietz, “Phorbol ester-induced apoptosis in prostate cancer cells via autocrine activation of the extrinsic apoptotic cascade: a key role for protein kinase Cδ,” The Journal of Biological Chemistry, vol. 280, no. 47, pp. 38982–38991, 2005.
[463]
V. A. von Burstin, L. Xiao, and M. G. Kazanietz, “Bryostatin 1 inhibits phorbol ester-induced apoptosis in prostate cancer cells by differentially modulating protein kinase C (PKC) δ translocation and preventing PKCδ-mediated release of tumor necrosis factor-α,” Molecular Pharmacology, vol. 78, no. 3, pp. 325–332, 2010.
[464]
L. Xiao, A. Gonzalez-Guerrico, and M. G. Kazanietz, “PKC-mediated secretion of death factors in LNCaP prostate cancer cells is regulated by androgens,” Molecular Carcinogenesis, vol. 48, no. 3, pp. 187–195, 2009.
[465]
L. Xiao, M. Eto, and M. G. Kazanietz, “ROCK mediates phorbol ester-induced apoptosis in prostate cancer cells via p2 up-regulation and JNK,” The Journal of Biological Chemistry, vol. 284, no. 43, pp. 29365–29375, 2009.
[466]
M. Sumitomo, R. Shen, J. S. Goldberg, J. Dai, D. Navarro, and D. M. Nanus, “Neutral endopeptidase promotes phorbol ester-induced apoptosis in prostate cancer cells by inhibiting neuropeptide-induced protein kinase C δ degradation,” Cancer Research, vol. 60, no. 23, pp. 6590–6596, 2000.
[467]
M. Sumitomo, M. Ohba, J. Asakuma et al., “Protein kinase Cδ amplifies ceramide formation via mitochondrial signaling in prostate cancer cells,” The Journal of Clinical Investigation, vol. 109, no. 6, pp. 827–836, 2002.
[468]
S. Kharait, R. Dhir, D. Lauffenburger, and A. Wells, “Protein kinase Cδ signaling downstream of the EGF receptor mediates migration and invasiveness of prostate cancer cells,” Biochemical and Biophysical Research Communications, vol. 343, no. 3, pp. 848–856, 2006.
[469]
H. S. Yu, T. H. Lin, and C. H. Tang, “Bradykinin enhances cell migration in human prostate cancer cells through B2 receptor/PKCδ/c-Src dependent signaling pathway,” Prostate, vol. 73, no. 1, pp. 89–100, 2013.
[470]
J. Kim, T. Koyanagi, and D. Mochly-Rosen, “PKCIδ activation mediates angiogenesis via NADPH oxidase activity in PC-3 prostate cancer cells,” Prostate, vol. 71, no. 9, pp. 946–954, 2011.
[471]
G. Kovacs, M. Akhtar, B. J. Beckwith et al., “The Heidelberg classification of renal cell tumours,” The Journal of Pathology, vol. 183, no. 2, pp. 131–133, 1997.
[472]
J. S. Lam, O. Shvarts, J. T. Leppert, R. A. Figlin, and A. S. Belldegrun, “Renal cell carcinoma 2005: new frontiers in staging, prognostication and targeted molecular therapy,” The Journal of Urology, vol. 173, no. 6, pp. 1853–1862, 2005.
[473]
M. von Brandenstein, J. J. Pandarakalam, L. Kroon et al., “MicroRNA 15a, inversely correlated to PKCα, is a potential marker to differentiate between benign and malignant renal tumors in biopsy and urine samples,” American Journal of Pathology, vol. 180, no. 5, pp. 1787–1797, 2012.
[474]
W. Brenner, F. Benzing, J. Gudejko-Thiel et al., “Regulation of β1 integrin expression by PKCε in renal cancer cells,” International Journal of Oncology, vol. 25, no. 4, pp. 1157–1163, 2004.
[475]
R. Engers, S. Mrzyk, E. Springer et al., “Protein kinase C in human renal cell carcinomas: role in invasion and differential isoenzyme expression,” British Journal of Cancer, vol. 82, no. 5, pp. 1063–1069, 2000.
[476]
W. Brenner, G. F?rber, T. Herget, C. Wiesner, J. G. Hengstler, and J. W. Thüroff, “Protein kinase C η is associated with progression of Renal Cell Carcinoma (RCC),” Anticancer Research, vol. 23, no. 5A, pp. 4001–4006, 2003.
[477]
Y. S. Pu, C. Y. Huang, J. Y. Chen, et al., “Down-regulation of PKCζ in renal cell carcinoma and its clinicopathological implications,” Journal of Biomedical Science, vol. 19, article 39, 2012.
[478]
K. Datta, J. Li, R. Bhattacharya, L. Gasparian, E. Wang, and D. Mukhopadhyay, “Protein kinase C ζ transactivates hypoxia-inducible factor α by promoting its association with p300 in renal cancer,” Cancer Research, vol. 64, no. 2, pp. 456–462, 2004.
[479]
B. Huang, K. Cao, X. Li et al., “The expression and role of protein kinase C (PKC) epsilon in clear cell renal cell carcinoma,” Journal of Experimental and Clinical Cancer Research, vol. 30, no. 1, article 88, 2011.
[480]
B. Zhan, C. Kong, K. Guo, and Z. Zhang, “PKCα is involved in the progression of kidney carcinoma through regulating netrin-1/UNC5B signaling pathway,” Tumor Biology, vol. 34, no. 3, pp. 1759–1766, 2013.
[481]
D. Lv, W. Zhao, D. Dong et al., “Genetic and epigenetic control of UNC5C expression in human renal cell carcinoma,” European Journal of Cancer, vol. 47, no. 13, pp. 2068–2076, 2011.
[482]
O. V. Razorenova, E. C. Finger, R. Colavitti et al., “VHL loss in renal cell carcinoma leads to up-regulation of CUB domain-containing protein 1 to stimulate PKCδ-driven migration,” Proceedings of the National Academy of Sciences of the United States of America, vol. 108, no. 5, pp. 1931–1936, 2011.
[483]
W. Brenner, I. Greber, J. Gudejko-Thiel et al., “Migration of renal carcinoma cells is dependent on protein kinase Cδ via β1 integrin and focal adhesion kinase,” International Journal of Oncology, vol. 32, no. 5, pp. 1125–1131, 2008.
[484]
K. Datta, R. Nambudripad, S. Pal, M. Zhou, H. T. Cohen, and D. Mukhopadhyay, “Inhibition of insulin-like growth factor-I-mediated cell signaling by the von Hippel-Lindau gene product in renal cancer,” The Journal of Biological Chemistry, vol. 275, no. 27, pp. 20700–20706, 2000.
[485]
N. Bergelin, C. L?f, S. Balthasar, V. Kalhori, and K. T?rnquist, “S1P1 and VEGFR-2 form a signaling complex with extracellularly regulated kinase 1/2 and protein kinase C-α regulating ML-1 thyroid carcinoma cell migration,” Endocrinology, vol. 151, no. 7, pp. 2994–3005, 2010.
[486]
N. Bergelin, T. Blom, J. Heikkil? et al., “Sphingosine kinase as an oncogene: autocrine sphingosine 1-phoshate modulates ML-1 thyroid carcinoma cell migration by a mechanism dependent on protein kinase C-α and ERK1/2,” Endocrinology, vol. 150, no. 5, pp. 2055–2063, 2009.
[487]
Y. Zhu, Q. Dong, B. J. Tan, W. G. Lim, S. Zhou, and W. Duan, “The PKCα-D294G mutant found in pituitary and thyroid tumors fails to transduce extracellular signals,” Cancer Research, vol. 65, no. 11, pp. 4520–4524, 2005.
[488]
C. Prévostel, V. Alvaro, F. de Boisvilliers, A. Martin, C. Jaffiol, and D. Joubert, “The natural protein kinase Cα mutant is present in human thyroid neoplasms,” Oncogene, vol. 11, no. 4, pp. 669–674, 1995.
[489]
C. Prévostel, A. Martin, V. Alvaro, C. Jaffiol, and D. Joubert, “Protein kinase C alpha and tumorigenesis of the endocrine gland,” Hormone Research, vol. 47, no. 4–6, pp. 140–144, 1997.
[490]
A. Vallentin, T.-C. Lo, and D. Joubert, “A single point mutation in the V3 region affects protein kinase Cα targeting and accumulation at cell-cell contacts,” Molecular and Cellular Biology, vol. 21, no. 10, pp. 3351–3363, 2001.
[491]
R. Assert, R. K?tter, U. Schiemann, P. Goretzki, and A. F. H. Pfeiffer, “Effects of the putatively oncogenic protein kinase Cα D294G mutation on enzymatic activity and cell growth and its occurrence in human thyroid neoplasias,” Hormone and Metabolic Research, vol. 34, no. 6, pp. 311–317, 2002.
[492]
J. A. Knauf, L. S. Ward, Y. E. Nikiforov et al., “Isozyme-specific abnormalities of PKC in thyroid cancer: evidence for post-transcriptional changes in PKC epsilon,” The Journal of Clinical Endocrinology and Metabolism, vol. 87, no. 5, pp. 2150–2159, 2002.
[493]
X. Zhang, D. Li, M. Li, et al., “MicroRNA-146a targets PRKCE to modulate papillary thyroid tumor development,” International Journal of Cancer, vol. 134, no. 2, pp. 257–267, 2014.
[494]
J. A. Knauf, R. Elisei, D. Mochly-Rosen et al., “Involvement of protein kinase Cε (PKCε) in thyroid cell death. A truncated chimeric PKCε cloned from a thyroid cancer cell line protects thyroid cells from apoptosis,” The Journal of Biological Chemistry, vol. 274, no. 33, pp. 23414–23425, 1999.
[495]
J. A. Knauf, B. Ouyang, M. Croyle, E. Kimura, and J. A. Fagin, “Acute expression of RET/PTC induces isozyme-specific activation and subsequent downregulation of PKCε in PCCL3 thyroid cells,” Oncogene, vol. 22, no. 44, pp. 6830–6838, 2003.
[496]
D. Molè, E. Gentilin, T. Gagliano et al., “Protein kinase C: a putative new target for the control of human medullary thyroid carcinoma cell proliferation in vitro,” Endocrinology, vol. 153, no. 5, pp. 2088–2098, 2012.
[497]
K. Koike, T. Fujii, A. M. Nakamura et al., “Activation of protein kinase C δ induces growth arrest in NPA thyroid cancer cells through extracellular signal-regulated kinase mitogen-activated protein kinase,” Thyroid, vol. 16, no. 4, pp. 333–341, 2006.
[498]
E. Afrasiabi, J. Ahlgren, N. Bergelin, and K. T?rnquist, “Phorbol 12-myristate 13-acetate inhibits FRO anaplastic human thyroid cancer cell proliferation by inducing cell cycle arrest in G1/S phase: evidence for an effect mediated by PKCδ,” Molecular and Cellular Endocrinology, vol. 292, no. 1-2, pp. 26–35, 2008.
[499]
N. Li, Z. X. Du, Z. H. Zong, et al., “PKCδ-mediated phosphorylation of BAG3 at Ser187 site induces epithelial-mesenchymal transition and enhances invasiveness in thyroid cancer FRO cells,” Oncogene, vol. 32, no. 88, pp. 4539–4548, 2013.
[500]
L. Zhang, J. Huang, N. Yang et al., “Integrative genomic analysis of protein kinase C (PKC) family identifies PKCι as a biomarker and potential oncogene in ovarian carcinoma,” Cancer Research, vol. 66, no. 9, pp. 4627–4635, 2006.
[501]
J. H.. Kang, T. Mori, H. Kitazaki, et al., “Kinase activity of protein kinase cα in serum as a diagnostic biomarker of human lung cancer,” Anticancer Research, vol. 33, no. 2, pp. 485–488, 2013.
[502]
J. H.. Kang, T. Mori, H. Kitazaki, et al., “Serum protein kinase Cα as a diagnostic biomarker of cancers,” Cancer Biomarkers, vol. 13, no. 2, pp. 99–103, 2013.
[503]
L. A. Davidson, C. M. Aymond, Y. Jiang, N. D. Turner, J. R. Lupton, and R. S. Chapkin, “Non-invasive detection of fecal protein kinase C βII and ζ messenger RNA: putative biomarkers for colon cancer,” Carcinogenesis, vol. 19, no. 2, pp. 253–257, 1998.
[504]
L. A. Davidson, Y. Jiang, J. R. Lupton, and R. S. Chapkin, “Noninvasive detection of putative biomarkers for colon cancer using fecal messenger RNA,” Cancer Epidemiology Biomarkers & Prevention, vol. 4, no. 6, pp. 643–647, 1995.
[505]
Q. Pan, L. W. Bao, C. G. Kleer et al., “Protein kinase Cε is a predictive biomarker of aggressive breast cancer and a validated target for RNA interference anticancer therapy,” Cancer Research, vol. 65, no. 18, pp. 8366–8371, 2005.
[506]
C. Cordon-Cardo, J. P. O'Brien, J. Boccia, D. Casals, J. R. Bertino, and M. R. Melamed, “Expression of the multidrug resistance gene product (P-glycoprotein) in human normal and tumor tissues,” Journal of Histochemistry and Cytochemistry, vol. 38, no. 9, pp. 1277–1287, 1990.
[507]
I. Sugawara, S. Akiyama, R. J. Scheper, and S. Itoyama, “Lung resistance protein (LRP) expression in human normal tissues in comparison with that of MDR1 and MRP,” Cancer Letters, vol. 112, no. 1, pp. 23–31, 1997.
[508]
D. S. Cox, K. R. Scott, H. Gao, and N. D. Eddington, “Effect of P-glycoprotein on the pharmacokinetics and tissue distribution of enaminone anticonvulsants: analysis by population and physiological approaches,” Journal of Pharmacology and Experimental Therapeutics, vol. 302, no. 3, pp. 1096–1104, 2002.
[509]
C. Sauvant, M. Nowak, C. Wirth et al., “Acidosis induces multi-drug resistance in rat prostate cancer cells (AT1) in vitro and in vivo by increasing the activity of the p-glycoprotein via activation of p38,” International Journal of Cancer, vol. 123, no. 11, pp. 2532–2542, 2008.
[510]
C. R. Carmo, J. Lyons-Lewis, M. J. Seckl, and A. P. Costa-Pereira, “A novel requirement for Janus Kinases as mediators of drug resistance induced by fibroblast growth factor-2 in human cancer cells,” PLoS ONE, vol. 6, no. 5, Article ID e19861, 2011.
[511]
S. Daenen, B. van der Holt, G. E. G. Verhoef et al., “Addition of cyclosporin A to the combination of mitoxantrone and etoposide to overcome resistance to chemotherapy in refractory or relapsing acute myeloid leukaemia: a randomised phase II trial from HOVON, the Dutch-Belgian Haemato-Oncology Working Group for adults,” Leukemia Research, vol. 28, no. 10, pp. 1057–1067, 2004.
[512]
M. R. Baer, S. L. George, R. K. Dodge et al., “Phase 3 study of the multidrug resistance modulator PSC-833 in previously untreated patients 60 years of age and older with acute myeloid leukemia: cancer and Leukemia Group B study 9720,” Blood, vol. 100, no. 4, pp. 1224–1232, 2002.
[513]
M. Hu, Y. Liu, C. Deng et al., “Enhanced invasiveness in multidrug resistant leukemic cells is associated with overexpression of P-glycoprotein and cellular inhibitor of apoptosis protein,” Leukemia & Lymphoma, vol. 52, no. 7, pp. 1302–1311, 2011.
[514]
T. Reya, S. J. Morrison, M. F. Clarke, and I. L. Weissman, “Stem cells, cancer, and cancer stem cells,” Nature, vol. 414, no. 6859, pp. 105–111, 2001.
[515]
L. V. Nguyen, R. Vanner, P. Dirks, and C. J. Eaves, “Cancer stem cells: an evolving concept,” Nature Reviews Cancer, vol. 12, no. 2, pp. 133–143, 2012.
[516]
L. Y. W. Bourguignon, C. Earle, G. Wong, C. C. Spevak, and K. Krueger, “Stem cell marker (Nanog) and Stat-3 signaling promote MicroRNA-21 expression and chemoresistance in hyaluronan/CD44-activated head and neck squamous cell carcinoma cells,” Oncogene, vol. 31, no. 2, pp. 149–160, 2012.
[517]
L. Y. W. Bourguignon, K. Peyrollier, W. Xia, and E. Gilad, “Hyaluronan-CD44 interaction activates stem cell marker Nanog, Stat-3-mediated MDR1 gene expression, and ankyrin-regulated multidrug efflux in breast and ovarian tumor cells,” The Journal of Biological Chemistry, vol. 283, no. 25, pp. 17635–17651, 2008.
[518]
L. Y. W. Bourguignon, G. Wong, C. Earle, and L. Chen, “Hyaluronan-CD44v3 interaction with Oct4-Sox2-Nanog promotes miR-302 expression leading to self-renewal, clonal formation, and cisplatin resistance in cancer stem cells from head and neck squamous cell carcinoma,” The Journal of Biological Chemistry, vol. 287, no. 39, pp. 32800–32824, 2012.
[519]
W. K. Chu, P. M. Dai, H. L. Ki, C. C. Pao, and J. K. Chen, “Nanog expression is negatively regulated by protein kinase C activities in human cancer cell lines,” Oncogene, vol. 34, no. 7, pp. 1497–1509, 2013.
[520]
A. Singh, A. Singh, J. M. Sand, E. Heninger, B. B. Hafeez, and A. K. Verma, “Protein kinase Cε, which is linked to ultraviolet radiation-induced development of squamous cell carcinomas, stimulates rapid turnover of adult hair follicle stem cells,” Journal of Skin Cancer, vol. 2013, Article ID 452425, 13 pages, 2013.
[521]
H. Harrison, G. Farnie, S. J. Howell et al., “Regulation of breast cancer stem cell activity by signaling through the Notch4 receptor,” Cancer Research, vol. 70, no. 2, pp. 709–718, 2010.
[522]
J. Yun, A. Pannuti, I. Espinoza, et al., “Crosstalk between PKCα and Notch-4 in endocrine-resistant breast cancer cells,” Oncogenesis, vol. 2, article e60, 2013.
[523]
W. L. Tam, H. Lu, J. Buikhuisen, et al., “Protein kinase C α is a central signaling node and therapeutic target for breast cancer stem cells,” Cancer Cell, vol. 24, no. 3, pp. 347–364, 2013.
[524]
B. L. J. Webb, S. J. Hirst, and M. A. Giembycz, “Protein kinase C isoenzymes: a review of their structure, regulation and role in regulating airways smooth muscle tone and mitogenesis,” British Journal of Pharmacology, vol. 130, no. 7, pp. 1433–1452, 2000.