File Download
  Links for fulltext
     (May Require Subscription)
Supplementary

Article: PDK1 promotes ovarian cancer metastasis by modulating tumor-mesothelial adhesion, invasion, and angiogenesis via α5β1 integrin and JNK/IL-8 signaling

TitlePDK1 promotes ovarian cancer metastasis by modulating tumor-mesothelial adhesion, invasion, and angiogenesis via α5β1 integrin and JNK/IL-8 signaling
Authors
KeywordsOxidoreductases
Pyruvate Kinase
Dichloroacetate DCA
Issue Date2020
PublisherSpringer Nature [academic journals on nature.com]: Fully open access journals. The Journal's web site is located at http://www.nature.com/oncsis/index.html
Citation
Oncogenesis, 2020, v. 9, p. article no. 24 How to Cite?
AbstractOvarian cancer is the most lethal gynecological malignancies owing to the lack of definitive symptoms until development of widespread metastases. Identification of novel prognostic and therapeutic targets is therefore an urgent need to improve survival. Here, we demonstrated high expression of the mitochondrial gatekeeping enzyme, pyruvate dehydrogenase kinase 1 (PDK1), in both clinical samples and cell lines of ovarian cancer. PDK1 expression was significantly associated with metastasis, reduced chemosensitivity, and poor overall and disease-free survival, and further highlighted as an independent prognostic factor. Silencing of PDK1 retarded lactate production, ovarian cancer cell adhesion, migration, invasion, and angiogenesis, and consequently metastasis, concomitant with decreased α5β1 integrin expression. Phospho-kinase array profiling and RNA sequencing analyses further revealed reduction of JNK activation and IL-8 expression in PDK1-depleted cells. Conversely, PDK1 overexpression promoted cell adhesion via modulation of α5β1 integrins, along with cell migration, invasion, and angiogenesis through activation of JNK/IL-8 signaling. PDK1 depletion additionally hindered tumor growth and dissemination in nude mice in vivo. Importantly, PDK1 levels were upregulated upon treatment with conditioned medium from omental tissues, which in turn promoted metastasis. Our findings suggest that PDK1, which is regulated by the tumor microenvironment, controls lactate production and promotes ovarian cancer cell metastasis via modulation of α5β1 integrin and JNK/IL-8 signaling. To our knowledge, this is the first report to demonstrate an association between PDK1 and survival in patients with ovarian cancer, supporting its efficacy as a valuable prognostic marker and therapeutic molecular target for the disease. Introd
Persistent Identifierhttp://hdl.handle.net/10722/281713
ISSN
2023 Impact Factor: 5.9
2023 SCImago Journal Rankings: 1.855
PubMed Central ID
ISI Accession Number ID

 

DC FieldValueLanguage
dc.contributor.authorSiu, MKY-
dc.contributor.authorJIANG, Y-X-
dc.contributor.authorWANG, J-J-
dc.contributor.authorLeung, THY-
dc.contributor.authorNgu, SF-
dc.contributor.authorCheung, ANY-
dc.contributor.authorNgan, HYS-
dc.contributor.authorChan, KKL-
dc.date.accessioned2020-03-22T04:18:38Z-
dc.date.available2020-03-22T04:18:38Z-
dc.date.issued2020-
dc.identifier.citationOncogenesis, 2020, v. 9, p. article no. 24-
dc.identifier.issn2157-9024-
dc.identifier.urihttp://hdl.handle.net/10722/281713-
dc.description.abstractOvarian cancer is the most lethal gynecological malignancies owing to the lack of definitive symptoms until development of widespread metastases. Identification of novel prognostic and therapeutic targets is therefore an urgent need to improve survival. Here, we demonstrated high expression of the mitochondrial gatekeeping enzyme, pyruvate dehydrogenase kinase 1 (PDK1), in both clinical samples and cell lines of ovarian cancer. PDK1 expression was significantly associated with metastasis, reduced chemosensitivity, and poor overall and disease-free survival, and further highlighted as an independent prognostic factor. Silencing of PDK1 retarded lactate production, ovarian cancer cell adhesion, migration, invasion, and angiogenesis, and consequently metastasis, concomitant with decreased α5β1 integrin expression. Phospho-kinase array profiling and RNA sequencing analyses further revealed reduction of JNK activation and IL-8 expression in PDK1-depleted cells. Conversely, PDK1 overexpression promoted cell adhesion via modulation of α5β1 integrins, along with cell migration, invasion, and angiogenesis through activation of JNK/IL-8 signaling. PDK1 depletion additionally hindered tumor growth and dissemination in nude mice in vivo. Importantly, PDK1 levels were upregulated upon treatment with conditioned medium from omental tissues, which in turn promoted metastasis. Our findings suggest that PDK1, which is regulated by the tumor microenvironment, controls lactate production and promotes ovarian cancer cell metastasis via modulation of α5β1 integrin and JNK/IL-8 signaling. To our knowledge, this is the first report to demonstrate an association between PDK1 and survival in patients with ovarian cancer, supporting its efficacy as a valuable prognostic marker and therapeutic molecular target for the disease. Introd-
dc.languageeng-
dc.publisherSpringer Nature [academic journals on nature.com]: Fully open access journals. The Journal's web site is located at http://www.nature.com/oncsis/index.html-
dc.relation.ispartofOncogenesis-
dc.rightsThis work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.-
dc.subjectOxidoreductases-
dc.subjectPyruvate Kinase-
dc.subjectDichloroacetate DCA-
dc.titlePDK1 promotes ovarian cancer metastasis by modulating tumor-mesothelial adhesion, invasion, and angiogenesis via α5β1 integrin and JNK/IL-8 signaling-
dc.typeArticle-
dc.identifier.emailSiu, MKY: mkysiu@hku.hk-
dc.identifier.emailNgu, SF: ngusiewf@hku.hk-
dc.identifier.emailCheung, ANY: anycheun@hkucc.hku.hk-
dc.identifier.emailNgan, HYS: hysngan@hkucc.hku.hk-
dc.identifier.emailChan, KKL: kklchan@hkucc.hku.hk-
dc.identifier.authoritySiu, MKY=rp00275-
dc.identifier.authorityNgu, SF=rp01367-
dc.identifier.authorityCheung, ANY=rp00542-
dc.identifier.authorityNgan, HYS=rp00346-
dc.identifier.authorityChan, KKL=rp00499-
dc.description.naturepublished_or_final_version-
dc.identifier.doi10.1038/s41389-020-0209-0-
dc.identifier.pmid32071289-
dc.identifier.pmcidPMC7028730-
dc.identifier.scopuseid_2-s2.0-85079733862-
dc.identifier.hkuros309441-
dc.identifier.volume9-
dc.identifier.spagearticle no. 24-
dc.identifier.epagearticle no. 24-
dc.identifier.isiWOS:000517272600001-
dc.publisher.placeUnited States-
dc.identifier.issnl2157-9024-

Export via OAI-PMH Interface in XML Formats


OR


Export to Other Non-XML Formats