Abstract

Protein kinase C ε (PKCε) has emerged as an oncogenic kinase and plays important roles in cell survival, mitogenesis and invasion. PKCε is up-regulated in most epithelial cancers, including prostate, breast, and lung cancer. Here we report that PKCε is an essential mediator of NF-κB activation in prostate cancer cells. A strong correlation exists between PKCε overexpression and NF-κB activation status in prostate cancer cells. Moreover, transgenic overexpression of PKCε in the mouse prostate causes preneoplastic lesions that display significant NF-κB hyperactivation. PKCε RNAi depletion or inhibition in prostate cancer cells diminishes NF-κB translocation to the nucleus with subsequent impairment of both activation of NF-κB transcription and induction of NF-κB responsive genes in response to the proinflammatory cytokine tumor necrosis factor α (TNFα). On the other hand, PKCε overexpression in normal prostate cells enhances activation of the NF-κB pathway. A mechanistic analysis revealed that TNFα activates PKCε via a C1 domain/diacylglycerol-dependent mechanism that involves phosphatidylcholine-phospholipase C. Moreover, PKCε facilitates the assembly of the TNF receptor-I signaling complex to trigger NF-κB activation. Our studies identified a molecular link between PKCε and NF-κB that controls key responses implicated in prostate cancer progression.

Highlights

  • PKC⑀, a potential oncogene, is up-regulated in prostate cancer

  • These results argue for a potential relationship between PKC⑀ and NF-␬B activation status in prostate cancer cells

  • Genetic deletion of the PKC⑀ gene inhibits the formation of prostate tumors in transgenic adenocarcinoma of mouse prostate (TRAMP) mice [51]

Read more

Summary

Background

PKC⑀, a potential oncogene, is up-regulated in prostate cancer. Results: PKC⑀ facilitates the formation of TNFR-I complex to regulate the NF-␬B pathway via a C1 domain/diacylglycerol-dependent mechanism. Binding of TNF␣ to its receptor TNFR-I in cancer cells, including prostate cancer cells, leads to the recruitment of adaptor proteins (TRADD, TRAF2, RIP) and the formation of a signaling complex that regulates NF-␬B activation and transcriptional activation of inflammatory, survival, and anti-apoptotic genes such as BCL2, BCL2L1, PTGS2 (COX2), and XIAP [3, 11,12,13]. Many studies highlighted the relevance of the atypical PKCs ␨ and ␫ as NF-␬B modulators [14, 15], Diacylglycerol (DAG)/phorbol ester responsive PKCs emerged as potential modifiers of NF-␬B signaling (16 –18) Both classical/conventional cPKCs (␣, ␤, and ␥) and novel nPKCs (␦, ⑀, ␩, and ␪) have been implicated as regulators of apoptosis, survival, differentiation, mitogenesis, and transformation in a strict cell-type dependent manner. Transgenic overexpression of PKC⑀ in mice conferred NF-␬B hyperactivation in preneoplastic lesions, arguing for a critical role for this nPKC in NF-␬B signaling

EXPERIMENTAL PROCEDURES
RESULTS
DISCUSSION
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.