File Download

Conference Paper: Function and modulation of Pdcd4 in ovarian cancer

TitleFunction and modulation of Pdcd4 in ovarian cancer
Authors
Issue Date2011
PublisherAmerican Association for Cancer Research.
Citation
The 102nd Annual Meeting of the American Association for Cancer Reseach (AACR 2011), Orlando, FL., 2-6 April 2011. How to Cite?
AbstractProgrammed cell death 4 (Pdcd4), a novel gene and originally identified as the neoplastic transformation inhibitor, is attenuated in various cancer types. It has been shown to have tumor suppressor properties such as inhibiting cell proliferation and inducing apoptosis. Our previous study demonstrated a continuous down-regulation of Pdcd4 expression in the sequence of normal-borderline-malignant ovarian tissue samples, and a significant correlation of Pdcd4 expression with disease-free survival. The objective of the current study is to further investigate the function and modulation of Pdcd4 in ovarian cancer cells. Our in vitro studies demonstrated that ectopic Pdcd4 expression significantly inhibited ovarian cancer cell proliferation by up-regulation of cyclin dependent kinase inhibitors of p27 and p21 and subsequently leading to cell cycle arrest at G1 stage. Unlike other cancers, Pdcd4 neither induce ovarian cancer cell apoptosis, nor promote the cellular sensitivity in response to cisplatin treatment. Pdcd4 over-expressing ovarian cancer cells exhibited elevated phosphatase and tensin homolog (PTEN) and decreased p-Akt expressions. In addition, Pdcd4 expression was up-regulated and translocated from cell nucleus to cytoplasm upon serum withdrawal treatment, but was rapidly depleted via proteasomal degradation upon serum re-administration. Treatment of a phosphoinositide 3-kinase (PI3K) inhibitor prevented the degradation of Pdcd4. In conclusion, loss of Pdcd4 may result in diminished cell ability to arrest at key checkpoint, leading to tumor development. The PI3K-Akt pathway may be involved in the regulation of Pdcd4 degradation in ovarian cancer cells. In response to the stress condition, Pdcd4 expression was altered and it was translocated to cytoplasm, suggesting that Pdcd4 was able to shuttle between cell compartments to perform its diverted functions.
DescriptionPoster Session 4 - Cytoplasmic Signal Transducers: abstract no. 1079
Persistent Identifierhttp://hdl.handle.net/10722/138088

 

DC FieldValueLanguage
dc.contributor.authorLiu, SSen_US
dc.contributor.authorWei, TNen_US
dc.contributor.authorChan, KKLen_US
dc.contributor.authorNgan, HYSen_US
dc.date.accessioned2011-08-26T14:40:03Z-
dc.date.available2011-08-26T14:40:03Z-
dc.date.issued2011en_US
dc.identifier.citationThe 102nd Annual Meeting of the American Association for Cancer Reseach (AACR 2011), Orlando, FL., 2-6 April 2011.en_US
dc.identifier.urihttp://hdl.handle.net/10722/138088-
dc.descriptionPoster Session 4 - Cytoplasmic Signal Transducers: abstract no. 1079-
dc.description.abstractProgrammed cell death 4 (Pdcd4), a novel gene and originally identified as the neoplastic transformation inhibitor, is attenuated in various cancer types. It has been shown to have tumor suppressor properties such as inhibiting cell proliferation and inducing apoptosis. Our previous study demonstrated a continuous down-regulation of Pdcd4 expression in the sequence of normal-borderline-malignant ovarian tissue samples, and a significant correlation of Pdcd4 expression with disease-free survival. The objective of the current study is to further investigate the function and modulation of Pdcd4 in ovarian cancer cells. Our in vitro studies demonstrated that ectopic Pdcd4 expression significantly inhibited ovarian cancer cell proliferation by up-regulation of cyclin dependent kinase inhibitors of p27 and p21 and subsequently leading to cell cycle arrest at G1 stage. Unlike other cancers, Pdcd4 neither induce ovarian cancer cell apoptosis, nor promote the cellular sensitivity in response to cisplatin treatment. Pdcd4 over-expressing ovarian cancer cells exhibited elevated phosphatase and tensin homolog (PTEN) and decreased p-Akt expressions. In addition, Pdcd4 expression was up-regulated and translocated from cell nucleus to cytoplasm upon serum withdrawal treatment, but was rapidly depleted via proteasomal degradation upon serum re-administration. Treatment of a phosphoinositide 3-kinase (PI3K) inhibitor prevented the degradation of Pdcd4. In conclusion, loss of Pdcd4 may result in diminished cell ability to arrest at key checkpoint, leading to tumor development. The PI3K-Akt pathway may be involved in the regulation of Pdcd4 degradation in ovarian cancer cells. In response to the stress condition, Pdcd4 expression was altered and it was translocated to cytoplasm, suggesting that Pdcd4 was able to shuttle between cell compartments to perform its diverted functions.-
dc.languageengen_US
dc.publisherAmerican Association for Cancer Research.-
dc.relation.ispartofAnnual Meeting of the American Association for Cancer Reseach, AACR 2011en_US
dc.titleFunction and modulation of Pdcd4 in ovarian canceren_US
dc.typeConference_Paperen_US
dc.identifier.emailLiu, SS: stephasl@hku.hken_US
dc.identifier.emailWei, TN: h0299984@HKUSUA.hku.hken_US
dc.identifier.emailChan, KKL: kklchan@hkucc.hku.hken_US
dc.identifier.emailNgan, HYS: gyonc@hku.hk-
dc.identifier.authorityLiu, SS=rp00372en_US
dc.identifier.authorityChan, KKL=rp00499en_US
dc.identifier.authorityNgan, HYS=rp00346en_US
dc.description.naturelink_to_OA_fulltext-
dc.identifier.hkuros190042en_US
dc.description.otherThe 102nd Annual Meeting of the American Association for Cancer Reseach (AACR 2011), Orlando, FL., 2-6 April 2011.-

Export via OAI-PMH Interface in XML Formats


OR


Export to Other Non-XML Formats