The unique protein kinase Cη: Implications for breast cancer (Review)

  • Authors:
    • Deepanwita Pal
    • Alakananda Basu
  • View Affiliations

  • Published online on: May 19, 2014     https://doi.org/10.3892/ijo.2014.2443
  • Pages: 493-498
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Deregulation of key signal transduction pathways that govern important cellular processes leads to cancer. The development of effective therapeutics for cancer warrants a comprehensive understanding of the signaling pathways that are deregulated in cancer. The protein kinase C (PKC) family has served as an attractive target for cancer therapy for decades owing to its crucial roles in several cellular processes. PKCη is a novel member of the PKC family that plays critical roles in various cellular processes such as growth, proliferation, differentiation and cell death. The regulation of PKCη appears to be unique compared to other PKC isozymes, and there are conflicting reports regarding its role in cancer. This review focuses on the unique aspects of PKCη in terms of its structure, regulation and subcellular distribution and speculates on how these features could account for its distinct functions. We have also discussed the functional implications of PKCη in cancer with particular emphasis on breast cancer.

1. Introduction

The protein kinase C (PKC) family is a family of serine/threonine kinases that play diverse roles in fundamental cellular processes including cell proliferation, cell death and differentiation (1,2). PKCs respond to extracellular signals that promote phospholipid hydrolysis and facilitate the generation of diacylglycerol (DAG) and release of Ca2+ from intracellular stores. These two second messengers activate PKCs in the presence of acidic phospholipids, such as phosphatidylserine (2). The PKC family garnered considerable attention by the discovery that PKCs could serve as receptors for tumor-promoting phorbol esters which was the first evidence to establish a link between PKCs and cancer (3,4). These phorbol esters are potent activators of PKCs and can substitute for the physiological stimulator DAG. Based on the structural features and cofactor requirements, the PKC family consists of 10 isozymes categorized as the conventional or the classical (c) PKCs (α, βI, βII, γ), the novel (n) PKCs (δ, ɛ, η, θ) and the atypical (a) PKCs (ζ, λ/ι) (1). While the conventional PKCs are sensitive to Ca2+ and DAG/phorbol ester, novel PKCs are insensitive to Ca2+ but respond to DAG/phorbol ester and atypical PKCs are insensitive to both Ca2+ and DAG/phorbol esters. The distinct structural and biochemical features of the PKC isozymes pave the way for the distinctive cellular responses attributed to the PKC family. Owing to the central role of PKCs in cellular regulation and signal transduction, significant research efforts have been devoted to the PKC family. However, much less is known about PKCη, which is a unique member of the novel PKC family. This review focuses on the biology of PKCη and its implications in breast cancer.

2. Protein kinase Cη, a unique PKC

Protein kinase Cη (PKCη) is a novel member of the PKC family. It is classified as a calcium-independent but DAG/phorbol ester-dependent PKC (5). It was first isolated from a cDNA library of mouse epidermis (5). PKCη is assigned to human chromosome 14 (14q22–23) and mouse chromosome 12 (12C3–D2) (6,7) and contains an open reading frame encoding 683 amino acid residues (8). Contrary to other PKCs which are primarily enriched in the brain tissue, PKCη is mainly expressed in lung, skin and heart tissues (9). PKCη participates in various cellular processes including proliferation, differentiation, secretion and apoptosis (1016). Recent reports have revealed the role of PKCη in immune function (17,18). PKCη was shown to be important for T-cell proliferation and homeostasis (19), and was also implicated in the regulation of toll-like receptor-2 (TLR-2) responses in macrophages (20).

3. Structure

All PKC isozymes contain a common structural backbone comprised of a highly conserved catalytic domain at the C-terminal and a regulatory domain at the N-terminal (Fig. 1). PKCs possess 4 conserved modules (C1–4): C1 and C2 are the membrane targeting modules that along with the pseudosubstrate region form the regulatory domain; C3 and C4 comprise the catalytic domain (21). A proteolytically labile hinge region connects the regulatory domain to the catalytic domain (22). The catalytic domain consists of motifs that are required for ATP/substrate binding and catalysis. The N-terminal contains the autoinhibitory pseudosubstrate sequence that contains an alanine in place of the serine/threonine phosphoacceptor site, but otherwise resembles a PKC substrate. The pseudosubstrate thus holds the enzyme in an inactive conformation by occupying the catalytic site (21). The pseudosubstrate sequence of PKCη is the most divergent amongst the PKC isozymes (9).

The structure of PKCη comprises of a highly conserved catalytic domain at the C-terminal and the regulatory domain at the N-terminal similar to other PKCs (21). A characteristic cysteine-rich region is present in the C1 domain of PKCη which allows binding to physiological stimulator DAG and pharmacological activators such as tumor-promoting phorbol esters (23). In addition, C1 domain confers selectivity for phosphatidylserine that acts as the activator for PKCs (24). The C2 domain of PKCη lacks the key aspartic acid residues to bind Ca2+ and consequently renders PKCη insensitive to Ca2+ (23). PKCη shares greatest homology with PKCɛ, another novel PKC (9).

Similar to other PKC isozymes, PKCη has three conserved phosphorylation sites - activation loop (Thr-513), turn motif (Thr-655) and hydrophobic domain (Ser-674) (21). Although the order of priming phosphorylations of PKCη is not well established, phosphoinositide-dependent kinase 1 (PDK1) is believed to phosphorylate PKCη at the activation loop in vitro (25). In mouse A9 fibroblasts infected with parovirus, Lachmann et al demonstrated that PKCλ phosphorylates PKCη at the hydrophobic site thus allowing PDK1 access to the activation loop (26). The C2 domain of PKCη was found to be similar to PKCɛ with significant differences at the putative lipid binding site. Mass spectrometric analysis of the C2 domain of PKCη revealed two autophosphorylation sites at Ser-28 and Ser-32 (27). The autophosphorylation site at Ser-28 but not Ser-32 is conserved in PKCɛ (27). It has been speculated that autophosphorylation at these sites could affect the lipid-binding of PKCη (27).

4. Regulation

The PKC isozymes are under tight structural and spatial regulation that underlies their biochemical functions, intracellular localization and tissue distribution (21). PKCs can be regulated by phosphorylation, cofactor binding and membrane targeting through interaction with scaffold proteins (28).

Anionic phospholipids such as phosphatidylserine and DAG/phorbol esters regulate PKCη (5,9). However, in contrast to other phorbol-ester sensitive PKC isozymes, PKCη resists downregulation by prolonged treatment with phorbol esters, suggesting its unique regulation (11,29,30). We have shown that PKCη not only resists downregulation by phorbol esters but is in fact upregulated by several structurally and functionally distinct PKC activators (31). We further reported that transphosphorylation by novel PKCɛ is responsible for activator-induced upregulation of PKCη (31).

PKCη is specifically activated by cholesterol sulfate and sulfatide (32). It was reported that cholesterol sulfate-mediated activation of PKCη involved casein kinase I (33). In addition, PKCη was shown to be activated by treatment with type I interferons (IFNs), IFNα or IFNβ in chronic myeloid leukemia cells (34). Interestingly, other novel PKC isozymes such as PKCδ, −ɛ and −θ are also activated by type I and type II IFNs and participate in type I and/or type II IFN-induced responses (3538). However, contrary to these isozymes, IFN-inducible transcription of IFN-stimulated genes or generation of antiviral responses is independent of PKCη. PKCη is also elevated in response to estradiol treatment in estrogen-sensitive breast cancer cells in a time- and concentration-dependent manner (39).

PKCη is subject to translational regulation under both normal and stressed conditions caused by amino acid starvation (40). Raveh-Amit and colleagues reported that the 5′-UTR of PKCη is unusually long (659 nucleotides) and rich in GC content and identified two upstream open reading frames (uORF) in the 5′-UTR which function as repressive elements under normal growth conditions. However, under amino acid starvation, the repression is removed by leaky scanning leading to the translational upregulation of PKCη (40). PKCɛ is the only other PKC isozyme for which the presence of a regulatory uORF has been reported (41).

5. Signal termination and downregulation of PKCη

Termination of PKC signaling can occur via different mechanisms such as release of PKC isozymes from the membrane, metabolism of DAG by DAG kinases (DGKs) (42,43), agonist-induced degradation or the removal of priming phosphorylation which leads to downregulation and rapid degradation (42,44,45). Several mechanisms of degradation have been proposed for the PKC isozymes. Conventional PKCs are believed to be downregulated by calcium-activated proteases, such as calpains (46,47) whereas PKCα, −δ and −ɛ were shown to be degraded via proteasome-mediated pathway (4850). Our studies have demonstrated that PKCη can be downregulated by both proteasomal-dependent and -independent pathways. While inhibition/knockdown of PDK1 caused PKCη downregulation via the proteasomal pathway, the downregulation of PKCη caused by the depletion of PKCɛ or by PKC inhibitors was independent of the proteasome-mediated pathway (51). Another study reported that dephosphorylation of PKCη was mediated by integrin-associated serine threonine phosphatase PP1γ in human platelets which was shown to be independent of the ubiquitin-mediated degradation (52). In addition, differential expression analysis in the neoplastic cell line 8701-BC demonstrated that PKCη downregulation can be induced by type V collagen (53).

6. Localization of PKCη

PKCη is localized in the Golgi, endoplasmic reticulum (ER) and the nuclear envelope (54). Although the C1A domain of PKCη lacks a Golgi localization signal similar to the other members of the novel PKC family, the C1B domain of PKCη facilitates its translocation to the Golgi complex (54). The localization of PKCη in the Golgi could account for the involvement of PKCη in Golgi vesicular transport. It has been previously reported that Golgi-cell surface transport requires protein kinase D (PKD) which is specifically activated by G protein subunits β1γ2 and β3γ2 via the Golgi-associated PKCη (55). In response to serum starvation and PMA, PKCη translocates to the nuclear envelope. While C1B domain is sufficient to drive Golgi translocation of PKCη, both the C1 and the pseudosubstrate region are required for the localization at the nuclear envelope and ER (54). PKCη is localized in the plasma membrane and the nuclear envelope upon stimulation with phorbol esters, while serum starvation leads to distribution only in the nuclear envelope (54). Furthermore, a recent study reported that in hepatocellular carcinoma cells, PKCη is targeted to lipid droplets where it limited the formation of larger lipid droplets (56).

7. Role of PKCη in breast cancer

PKC isozymes have been extensively researched as potent targets for cancer therapeutics since their discovery as receptors for tumor promoters (3,57). The role of PKCη in cancer is controversial owing to its divergent responses in different cancers. Although, PKCη-deficient mice were more susceptible to tumor promotion in two-stage skin carcinogenesis model (58), PKCη mediates chemotherapeutic resistance in breast cancer (10,59), glioblastoma (60), lung cancer (61) and several other cancers (62,63). It has been reported that PKCη is downregulated in hepatocellular carcinoma (64) but is associated with the progression of renal cell carcinoma (65). Thus, PKCη may promote or inhibit malignant growth depending on the cellular context.

PKCη is a regulator of mammary gland development (66). It is upregulated in the rat mammary gland during the transition from the resting to the pregnant state (66). Furthermore, a marked decrease in PKCη levels was observed during gland regression which is typically characterized by the onset of apoptotic processes leading to involution (66). Qualitative and quantitative alterations in PKCη have been reported in human breast cancer tissues (67). PKCη expression was increased in locally invasive breast tumor tissues and high levels of PKCη were detected in invasive tumors associated with significant lymph node metastases which suggests a role for PKCη in cancer progression (67). This is consistent with a report which demonstrated the importance of PKCη in maintaining tight junction integrity via interaction and subsequent phosphorylation of occludin on its C-terminal domain (68). Since key changes in the barrier function of tight junctions have been shown to be critical in cancer progression (69), it is likely that PKCη may have potential roles in survival and progression of cancer cells. We also observed that the levels of PKCη progressively increase with breast cancer aggressiveness in the MCF10 breast cancer series (51). It is also noteworthy that the promoter region of PKCη contains multiple sites for the transcription factors Ets1 and AP-1 (6), both of which have been implicated in breast cancer growth and progression (70,71). However, contrary to these reports, decreased PKCη expression was observed in invasive breast tumor tissues compared to the surrounding normal epithelium, suggesting that PKCη is decreased during breast cancer progression (67). Thus the role of PKCη in cancer progression remains controversial.

PKCη mRNA is elevated in multidrug-resistant breast tumors (72), and overexpression of PKCη has been shown to protect against apoptosis (10,11,15). We have previously reported that overexpression of PKCη attenuated caspase activation and TNF-induced cell death in breast cancer cells (10). PKCη also protects against camptothecin-induced DNA damage by activating NF-κB and promoting nuclear localization of RelA/p65 in breast cancer (15). Upon etoposide-induced stress, PKCη is tethered to the nuclear membrane and confers protection against cell death (73). Moreover, PKCη was effective in blocking apoptosis via the suppression of c-Jun N-terminal kinase (JNK) activity upon UV irradiation (59). PKCη is also critical for cell cycle control. Although PKCη induced growth arrest in NIH3T3 fibroblasts and keratinocytes (74,75), it enhanced cell cycle progression in breast cancer cells (12). Induced expression of PKCη led to an increase in the levels of cyclin E and cyclin D (12). Although the levels of the cell cycle inhibitor p27 (kip1) were unaltered by PKCη overexpression, it facilitated the removal of the cell cycle inhibitor p27 (kip1) from the cyclin E/cdk2 complex, thereby activating the cyclin E/cdk2 complex (12). Consistent with these findings, we observed that PKCη promotes breast cancer cell growth and proliferation, similar to its role in glioblastoma (14,51,76). On the other hand, PKCη was shown to negatively regulate Akt leading to decrease in insulin-like growth factor I (IGF-I)-induced cell proliferation in MCF-7 breast cancer cells (77). There are thus, contrasting functional responses of PKCη not only in different cancers, but also within the same cancer type.

8. Conclusions

PKCη is a unique member of the PKC family. Its distinct regulation in response to tumor promoters compared to the other PKCs has potential implications in cancer. Although several studies have established the role of PKCη in cell growth, proliferation and chemoresistance, conflicting reports have added ambiguity to the functional role of PKCη. Moreover, PKCη interacts with several signaling pathways, such as the PI3K/Akt, NF-κB and ERK/Elk-1 (15,76,78). In addition, most cells express multiple PKC isozymes which display redundant as well as opposing functions. The distinct biochemical properties, tissue distribution and subcellular localization of different PKC isozymes have been reported to result in divergent responses in cancer (7981). Thus, the crosstalk between these proteins will eventually influence the final outcome.

While the published reports have helped discern the regulation and function of PKCη, many questions remain regarding the paradoxical actions of PKCη. It would be worthwhile to understand the specific interactions of PKCη with other signaling pathways and the subsequent consequences on cellular regulation. Studies focused on the interaction of transcription factors such as AP-1 or Ets1 with PKCη in breast cancer could also yield interesting results. Thus, future studies should help determine the molecular cues which govern the dynamic role of PKCη.

Acknowledgements

This work was supported by the Doctoral Bridge Funding Award (DP) from the University of North Texas Health Science Center.

References

1 

Nishizuka Y: Intracellular signaling by hydrolysis of phospholipids and activation of protein kinase C. Science. 258:607–614. 1992. View Article : Google Scholar : PubMed/NCBI

2 

Griner EM and Kazanietz MG: Protein kinase C and other diacylglycerol effectors in cancer. Nat Rev Cancer. 7:281–294. 2007. View Article : Google Scholar : PubMed/NCBI

3 

Blumberg PM: Protein kinase C as the receptor for the phorbol ester tumor promoters: sixth Rhoads memorial award lecture. Cancer Res. 48:1–8. 1988.PubMed/NCBI

4 

Castagna M, Takai Y, Kaibuchi K, Sano K, Kikkawa U and Nishizuka Y: Direct activation of calcium-activated, phospholipid-dependent protein kinase by tumor-promoting phorbol esters. J Biol Chem. 257:7847–7851. 1982.PubMed/NCBI

5 

Osada S, Mizuno K, Saido TC, et al: A phorbol ester receptor/protein kinase, nPKC eta, a new member of the protein kinase C family predominantly expressed in lung and skin. J Biol Chem. 265:22434–22440. 1990.PubMed/NCBI

6 

Quan T and Fisher GJ: Cloning and characterization of the human protein kinase C-eta promoter. J Biol Chem. 274:28566–28574. 1999. View Article : Google Scholar : PubMed/NCBI

7 

Chida K, Nakada T, Otsuka H, Kuroki T and Satoh H: Assignment of protein kinase C eta (Pkch) to mouse chromosome band 12C3–D2 by in situ hybridization. Cytogenet Cell Genet. 82:30–31. 1998.PubMed/NCBI

8 

Kashiwagi M, Ohba M, Chida K and Kuroki T: Protein kinase C eta (PKC eta): its involvement in keratinocyte differentiation. J Biochem. 132:853–857. 2002. View Article : Google Scholar : PubMed/NCBI

9 

Bacher N, Zisman Y, Berent E and Livneh E: Isolation and characterization of PKC-L, a new member of the protein kinase C-related gene family specifically expressed in lung, skin, and heart. Mol Cell Biol. 11:126–133. 1991.

10 

Akkaraju GR and Basu A: Overexpression of protein kinase C-eta attenuates caspase activation and tumor necrosis factor-alpha-induced cell death. Biochem Biophys Res Commun. 279:103–107. 2000. View Article : Google Scholar : PubMed/NCBI

11 

Basu A: The involvement of novel protein kinase C isozymes in influencing sensitivity of breast cancer MCF-7 cells to tumor necrosis factor-alpha. Mol Pharmacol. 53:105–111. 1998.PubMed/NCBI

12 

Fima E, Shtutman M, Libros P, et al: PKCeta enhances cell cycle progression, the expression of G1 cyclins and p21 in MCF-7 cells. Oncogene. 20:6794–6804. 2001. View Article : Google Scholar : PubMed/NCBI

13 

Fima E, Shahaf G, Hershko T, Apte RN and Livneh E: Expression of PKCeta in NIH-3T3 cells promotes production of the pro-inflammatory cytokine interleukin-6. Eur Cytokine Netw. 10:491–500. 1999.

14 

Hussaini IM, Karns LR, Vinton G, et al: Phorbol 12-myristate 13-acetate induces protein kinase ceta-specific proliferative response in astrocytic tumor cells. J Biol Chem. 275:22348–22354. 2000. View Article : Google Scholar : PubMed/NCBI

15 

Raveh-Amit H, Hai N, Rotem-Dai N, Shahaf G, Gopas J and Livneh E: Protein kinase Ceta activates NF-kappaB in response to camptothecin-induced DNA damage. Biochem Biophys Res Commun. 412:313–317. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Ohba M, Ishino K, Kashiwagi M, et al: Induction of differentiation in normal human keratinocytes by adenovirus-mediated introduction of the eta and delta isoforms of protein kinase C. Mol Cell Biol. 18:5199–5207. 1998.PubMed/NCBI

17 

Fu G and Gascoigne NR: Protein kinase Ceta, an emerging player in T-cell biology. Cell Cycle. 11:837–838. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Quann EJ, Liu X, Altan-Bonnet G and Huse M: A cascade of protein kinase C isozymes promotes cytoskeletal polarization in T cells. Nat Immunol. 12:647–654. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Fu G, Hu J, Niederberger-Magnenat N, et al: Protein kinase C eta is required for T cell activation and homeostatic proliferation. Sci Signal. 4:ra842011.PubMed/NCBI

20 

Park DW, Lee HK, Lyu JH, et al: TLR2 stimulates ABCA1 expression via PKC-eta and PLD2 pathway. Biochem Biophys Res Commun. 430:933–937. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Steinberg SF: Structural basis of protein kinase C isoform function. Physiol Rev. 88:1341–1378. 2008. View Article : Google Scholar : PubMed/NCBI

22 

Inoue M, Kishimoto A, Takai Y and Nishizuka Y: Studies on a cyclic nucleotide-independent protein kinase and its proenzyme in mammalian tissues. II Proenzyme and its activation by calcium-dependent protease from rat brain. J Biol Chem. 252:7610–7616. 1977.PubMed/NCBI

23 

Hashimoto Y, Osada S, Ohno S and Kuroki T: A Ca(2+)-independent protein kinase C, nPKC eta: its structure, distribution and possible function. Tohoku J Exp Med. 168:275–278. 1992. View Article : Google Scholar : PubMed/NCBI

24 

Johnson JE, Giorgione J and Newton AC: The C1 and C2 domains of protein kinase C are independent membrane targeting modules, with specificity for phosphatidylserine conferred by the C1 domain. Biochemistry. 39:11360–11369. 2000. View Article : Google Scholar : PubMed/NCBI

25 

Balendran A, Hare GR, Kieloch A, Williams MR and Alessi DR: Further evidence that 3-phosphoinositide-dependent protein kinase-1 (PDK1) is required for the stability and phosphorylation of protein kinase C (PKC) isoforms. FEBS Lett. 484:217–223. 2000. View Article : Google Scholar : PubMed/NCBI

26 

Lachmann S, Bar S, Rommelaere J and Nuesch JP: Parvovirus interference with intracellular signalling: mechanism of PKCeta activation in MVM-infected A9 fibroblasts. Cell Microbiol. 10:755–769. 2008. View Article : Google Scholar : PubMed/NCBI

27 

Littler DR, Walker JR, She YM, Finerty PJ Jr, Newman EM and Dhe-Paganon S: Structure of human protein kinase C eta (PKCeta) C2 domain and identification of phosphorylation sites. Biochem Biophys Res Commun. 349:1182–1189. 2006. View Article : Google Scholar : PubMed/NCBI

28 

Gould CM and Newton AC: The life and death of protein kinase C. Curr Drug Targets. 9:614–625. 2008. View Article : Google Scholar : PubMed/NCBI

29 

Chen CC, Wang JK and Chen WC: TPA induces translocation but not down-regulation of new PKC isoform eta in macrophages, MDCK cells and astrocytes. FEBS Lett. 412:30–34. 1997. View Article : Google Scholar : PubMed/NCBI

30 

Resnick MS, Luo X, Vinton EG and Sando JJ: Selective up-regulation of protein kinase C eta in phorbol ester-sensitive versus-resistant EL4 mouse thymoma cells. Cancer Res. 57:2209–2215. 1997.PubMed/NCBI

31 

Pal D, Outram SP and Basu A: Novel regulation of protein kinase C-eta. Biochem Biophys Res Commun. 425:836–841. 2012. View Article : Google Scholar

32 

Chida K, Murakami A, Tagawa T, Ikuta T and Kuroki T: Cholesterol sulfate, a second messenger for the eta isoform of protein kinase C, inhibits promotional phase in mouse skin carcinogenesis. Cancer Res. 55:4865–4869. 1995.PubMed/NCBI

33 

Okano M, Yokoyama T, Miyanaga T and Ohtsuki K: Activation of C-kinase eta through its cholesterol-3-sulfate-dependent phosphorylation by casein kinase I in vitro. Biol Pharm Bull. 27:109–112. 2004. View Article : Google Scholar : PubMed/NCBI

34 

Redig AJ, Sassano A, Majchrzak-Kita B, et al: Activation of protein kinase C{eta} by type I interferons. J Biol Chem. 284:10301–10314. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Uddin S, Sassano A, Deb DK, et al: Protein kinase C-delta (PKC-delta) is activated by type I interferons and mediates phosphorylation of Stat1 on serine 727. J Biol Chem. 277:14408–14416. 2002. View Article : Google Scholar : PubMed/NCBI

36 

Srivastava KK, Batra S, Sassano A, et al: Engagement of protein kinase C-theta in interferon signaling in T-cells. J Biol Chem. 279:29911–29920. 2004. View Article : Google Scholar : PubMed/NCBI

37 

Ivaska J, Bosca L and Parker PJ: PKCepsilon is a permissive link in integrin-dependent IFN-gamma signalling that facilitates JAK phosphorylation of STAT1. Nat Cell Biol. 5:363–369. 2003. View Article : Google Scholar : PubMed/NCBI

38 

Venkatesan BA, Mahimainathan L, Ghosh-Choudhury N, et al: PI 3 kinase-dependent Akt kinase and PKCepsilon independently regulate interferon-gamma-induced STAT1alpha serine phosphorylation to induce monocyte chemotactic protein-1 expression. Cell Signal. 18:508–518. 2006. View Article : Google Scholar

39 

Karp G, Maissel A and Livneh E: Hormonal regulation of PKC: estrogen up-regulates PKCeta expression in estrogen-responsive breast cancer cells. Cancer Lett. 246:173–181. 2007. View Article : Google Scholar : PubMed/NCBI

40 

Raveh-Amit H, Maissel A, Poller J, et al: Translational control of protein kinase Ceta by two upstream open reading frames. Mol Cell Biol. 29:6140–6148. 2009. View Article : Google Scholar : PubMed/NCBI

41 

Morrish BC and Rumsby MG: The 5′ UTR of protein kinase C epsilon confers translational regulation in vitro and in vivo. Biochem Biophys Res Commun. 283:1091–1098. 2001.

42 

Feng X, Zhang J, Barak LS, Meyer T, Caron MG and Hannun YA: Visualization of dynamic trafficking of a protein kinase C betaII/green fluorescent protein conjugate reveals differences in G protein-coupled receptor activation and desensitization. J Biol Chem. 273:10755–10762. 1998. View Article : Google Scholar

43 

Crotty T, Cai J, Sakane F, Taketomi A, Prescott SM and Topham MK: Diacylglycerol kinase delta regulates protein kinase C and epidermal growth factor receptor signaling. Proc Natl Acad Sci USA. 103:15485–15490. 2006. View Article : Google Scholar : PubMed/NCBI

44 

Leontieva OV and Black JD: Identification of two distinct pathways of protein kinase Calpha down-regulation in intestinal epithelial cells. J Biol Chem. 279:5788–5801. 2004. View Article : Google Scholar : PubMed/NCBI

45 

Newton AC: Protein kinase C: poised to signal. Am J Physiol Endocrinol Metab. 298:E395–E402. 2010. View Article : Google Scholar : PubMed/NCBI

46 

Pontremoli S, Melloni E, Damiani G, et al: Effects of a monoclonal anti-calpain antibody on responses of stimulated human neutrophils. Evidence for a role for proteolytically modified protein kinase C. J Biol Chem. 263:1915–1919. 1988.PubMed/NCBI

47 

Savart M, Letard P, Bultel S and Ducastaing A: Induction of protein kinase C down-regulation by the phorbol ester TPA in a calpain/protein kinase C complex. Int J Cancer. 52:399–403. 1992. View Article : Google Scholar : PubMed/NCBI

48 

Lee HW, Smith L, Pettit GR and Smith JB: Bryostatin 1 and phorbol ester down-modulate protein kinase C-alpha and -epsilon via the ubiquitin/proteasome pathway in human fibroblasts. Mol Pharmacol. 51:439–447. 1997.PubMed/NCBI

49 

Lee HW, Smith L, Pettit GR, Vinitsky A and Smith JB: Ubiquitination of protein kinase C-alpha and degradation by the proteasome. J Biol Chem. 271:20973–20976. 1996. View Article : Google Scholar : PubMed/NCBI

50 

Lu Z, Liu D, Hornia A, Devonish W, Pagano M and Foster DA: Activation of protein kinase C triggers its ubiquitination and degradation. Mol Cell Biol. 18:839–845. 1998.PubMed/NCBI

51 

Pal D, Outram SP and Basu A: Upregulation of PKCeta by PKCepsilon and PDK1 involves two distinct mechanisms and promotes breast cancer cell survival. Biochim Biophys Acta. 1830:4040–4045. 2013. View Article : Google Scholar

52 

Bynagari YS, Nagy B Jr, Tuluc F, et al: Mechanism of activation and functional role of protein kinase Ceta in human platelets. J Biol Chem. 284:13413–13421. 2009. View Article : Google Scholar

53 

Luparello C, Sirchia R and Longo A: Type V collagen and protein kinase C eta down-regulation in 8701-BC breast cancer cells. Mol Carcinog. 52:348–358. 2013. View Article : Google Scholar : PubMed/NCBI

54 

Maissel A, Marom M, Shtutman M, Shahaf G and Livneh E: PKCeta is localized in the Golgi, ER and nuclear envelope and translocates to the nuclear envelope upon PMA activation and serum-starvation: C1b domain and the pseudosubstrate containing fragment target PKCeta to the Golgi and the nuclear envelope. Cell Signal. 18:1127–1139. 2006. View Article : Google Scholar

55 

Diaz Anel AM and Malhotra V: PKCeta is required for beta1gamma2/beta3gamma2- and PKD-mediated transport to the cell surface and the organization of the Golgi apparatus. J Cell Biol. 169:83–91. 2005.PubMed/NCBI

56 

Suzuki M, Iio Y, Saito N and Fujimoto T: Protein kinase Ceta is targeted to lipid droplets. Histochem Cell Biol. 139:505–511. 2013. View Article : Google Scholar

57 

Mackay HJ and Twelves CJ: Targeting the protein kinase C family: are we there yet? Nat Rev Cancer. 7:554–562. 2007. View Article : Google Scholar : PubMed/NCBI

58 

Chida K, Hara T, Hirai T, et al: Disruption of protein kinase Ceta results in impairment of wound healing and enhancement of tumor formation in mouse skin carcinogenesis. Cancer Res. 63:2404–2408. 2003.PubMed/NCBI

59 

Rotem-Dai N, Oberkovitz G, Abu-Ghanem S and Livneh E: PKCeta confers protection against apoptosis by inhibiting the pro-apoptotic JNK activity in MCF-7 cells. Exp Cell Res. 315:2616–2623. 2009. View Article : Google Scholar : PubMed/NCBI

60 

Hussaini IM, Carpenter JE, Redpath GT, Sando JJ, Shaffrey ME and Vandenberg SR: Protein kinase C-eta regulates resistance to UV- and gamma-irradiation-induced apoptosis in glioblastoma cells by preventing caspase-9 activation. Neuro Oncol. 4:9–21. 2002. View Article : Google Scholar : PubMed/NCBI

61 

Sonnemann J, Gekeler V, Ahlbrecht K, et al: Down-regulation of protein kinase Ceta by antisense oligonucleotides sensitises A549 lung cancer cells to vincristine and paclitaxel. Cancer Lett. 209:177–185. 2004. View Article : Google Scholar : PubMed/NCBI

62 

Sonnemann J, Gekeler V, Sagrauske A, Muller C, Hofmann HP and Beck JF: Down-regulation of protein kinase Ceta potentiates the cytotoxic effects of exogenous tumor necrosis factor-related apoptosis-inducing ligand in PC-3 prostate cancer cells. Mol Cancer Ther. 3:773–781. 2004.

63 

Abu-Ghanem S, Oberkovitz G, Benharroch D, Gopas J and Livneh E: PKCeta expression contributes to the resistance of Hodgkin’s lymphoma cell lines to apoptosis. Cancer Biol Ther. 6:1375–1380. 2007.PubMed/NCBI

64 

Lu HC, Chou FP, Yeh KT, Chang YS, Hsu NC and Chang JG: Analysing the expression of protein kinase C eta in human hepatocellular carcinoma. Pathology. 41:626–629. 2009. View Article : Google Scholar : PubMed/NCBI

65 

Brenner W, Farber G, Herget T, Wiesner C, Hengstler JG and Thuroff JW: Protein kinase C eta is associated with progression of renal cell carcinoma (RCC). Anticancer Res. 23:4001–4006. 2003.PubMed/NCBI

66 

Masso-Welch PA, Verstovsek G, Darcy K, Tagliarino C and Ip MM: Protein kinase C eta upregulation and secretion during postnatal rat mammary gland differentiation. Eur J Cell Biol. 77:48–59. 1998. View Article : Google Scholar : PubMed/NCBI

67 

Masso-Welch PA, Winston JS, Edge S, et al: Altered expression and localization of PKC eta in human breast tumors. Breast Cancer Res Treat. 68:211–223. 2001. View Article : Google Scholar : PubMed/NCBI

68 

Suzuki T, Elias BC, Seth A, et al: PKC eta regulates occludin phosphorylation and epithelial tight junction integrity. Proc Natl Acad Sci USA. 106:61–66. 2009. View Article : Google Scholar : PubMed/NCBI

69 

Martin TA, Mason MD and Jiang WG: Tight junctions in cancer metastasis. Front Biosci (Landmark Ed). 16:898–936. 2011. View Article : Google Scholar : PubMed/NCBI

70 

Lincoln DW II and Bove K: The transcription factor Ets-1 in breast cancer. Front Biosci. 10:506–511. 2005. View Article : Google Scholar : PubMed/NCBI

71 

Shen Q, Uray IP, Li Y, et al: The AP-1 transcription factor regulates breast cancer cell growth via cyclins and E2F factors. Oncogene. 27:366–377. 2008. View Article : Google Scholar : PubMed/NCBI

72 

Beck J, Bohnet B, Brugger D, et al: Multiple gene expression analysis reveals distinct differences between G2 and G3 stage breast cancers, and correlations of PKC eta with MDR1, MRP and LRP gene expression. Br J Cancer. 77:87–91. 1998. View Article : Google Scholar : PubMed/NCBI

73 

Tamarkin A, Zurgil U, Braiman A, et al: DNA damage targets PKCeta to the nuclear membrane via its C1b domain. Exp Cell Res. 317:1465–1475. 2011. View Article : Google Scholar : PubMed/NCBI

74 

Cabodi S, Calautti E, Talora C, Kuroki T, Stein PL and Dotto GP: A PKC-eta/Fyn-dependent pathway leading to keratinocyte growth arrest and differentiation. Mol Cell. 6:1121–1129. 2000. View Article : Google Scholar : PubMed/NCBI

75 

Livneh E, Shimon T, Bechor E, Doki Y, Schieren I and Weinstein IB: Linking protein kinase C to the cell cycle: ectopic expression of PKC eta in NIH3T3 cells alters the expression of cyclins and Cdk inhibitors and induces adipogenesis. Oncogene. 12:1545–1555. 1996.PubMed/NCBI

76 

Uht RM, Amos S, Martin PM, Riggan AE and Hussaini IM: The protein kinase C-eta isoform induces proliferation in glioblastoma cell lines through an ERK/Elk-1 pathway. Oncogene. 26:2885–2893. 2007. View Article : Google Scholar : PubMed/NCBI

77 

Shahaf G, Rotem-Dai N, Koifman G, Raveh-Amit H, Frost SA and Livneh E: PKCeta is a negative regulator of AKT inhibiting the IGF-I induced proliferation. Exp Cell Res. 318:789–799. 2012. View Article : Google Scholar : PubMed/NCBI

78 

Aeder SE, Martin PM, Soh JW and Hussaini IM: PKC-eta mediates glioblastoma cell proliferation through the Akt and mTOR signaling pathways. Oncogene. 23:9062–9069. 2004. View Article : Google Scholar : PubMed/NCBI

79 

Koivunen J, Aaltonen V and Peltonen J: Protein kinase C (PKC) family in cancer progression. Cancer Lett. 235:1–10. 2006. View Article : Google Scholar : PubMed/NCBI

80 

Urtreger AJ, Kazanietz MG and Bal de Kier Joffe ED: Contribution of individual PKC isoforms to breast cancer progression. IUBMB Life. 64:18–26. 2012. View Article : Google Scholar : PubMed/NCBI

81 

Basu A and Pal D: Two faces of protein kinase Cdelta: the contrasting roles of PKCdelta in cell survival and cell death. Sci World J. 10:2272–2284. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2014
Volume 45 Issue 2

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Pal D and Pal D: The unique protein kinase Cη: Implications for breast cancer (Review). Int J Oncol 45: 493-498, 2014
APA
Pal, D., & Pal, D. (2014). The unique protein kinase Cη: Implications for breast cancer (Review). International Journal of Oncology, 45, 493-498. https://doi.org/10.3892/ijo.2014.2443
MLA
Pal, D., Basu, A."The unique protein kinase Cη: Implications for breast cancer (Review)". International Journal of Oncology 45.2 (2014): 493-498.
Chicago
Pal, D., Basu, A."The unique protein kinase Cη: Implications for breast cancer (Review)". International Journal of Oncology 45, no. 2 (2014): 493-498. https://doi.org/10.3892/ijo.2014.2443