MIEN1 Drives Breast Cancer Invasion by Regulating Cytoskeletal-Focal Adhesions Dynamics

dc.contributor.advisorVishwanatha, Jamboor K.
dc.contributor.committeeMemberClark, Abbot F.
dc.contributor.committeeMemberBasu, Alakananda
dc.creatorKpetemey, Marilyne F.
dc.date.accessioned2019-08-22T21:09:33Z
dc.date.available2019-08-22T21:09:33Z
dc.date.issued2015-05-01
dc.date.submitted2015-07-08T08:36:33-07:00
dc.description.abstractIn the recent years, Migration and Invasion Enhancer 1(MIEN1) has emerged as a potential biomarker and a plausible target in breast cancer. Located in the 17q12-21 region of the human chromosome, next to the Her-2/neu loci, MIEN1 presents a robust expression in breast carcinomas; however is completely absent or low in the normal tissues. MIEN1 is post-translationally modified by geranyl-geranyl transferase-I (GgtaseI), which adds isoprenyl group to the carboxyl-terminal of the protein. Prenylated MIEN1 then associates with the inner leaflet of the plasma membrane and acts as an adaptor protein triggering downstream signaling through the Akt/NF-kB axis to regulate the expression of key proteases and angiogenic factors like MMP-9, uPA and VEGF. In migrating cells, MIEN1 enhances filopodium formation at the leading edge. Aside from its prenylation and redox-active motifs, MIEN1 also contains a canonical ITAM, reported to be associated with epithelial-to-mesenchymal transition. Although the role MIEN1 in cell migration and invasion is well known, the underlying molecular mechanisms remain elusive. Here, we show that MIEN1 interacts with Annexin A2, a cytoskeletal protein and a regulator of the plasminogen/plasmin system in breast cancer cells to increase migration and invasion. We confirmed that MIEN1 regulates actin dynamics by associating with cytoskeletal effectors in the lamellum. We also show that MIEN1 expression redirects breast tumor cell migration toward a collective migration. Our studies validate MIEN1-ITAM and CAAX as key motifs to MIEN1-induced functions. In conclusion, our findings confirm the role of MIEN1 in the remodeling of the actin cytoskeleton during motility. Furthermore it attests to previous findings suggesting that motility patterns depend on various environmental factors along with regulatory genes involved. Our study demonstrates an interesting example from cell biology where adaptor proteins regulate various signaling pathways and control cellular processes through protein-protein interactions.
dc.format.mimetypeapplication/pdf
dc.identifier.urihttps://hdl.handle.net/20.500.12503/29116
dc.language.isoen
dc.provenance.legacyDownloads6
dc.subjectBiological Phenomena, Cell Phenomena, and Immunity
dc.subjectCancer Biology
dc.subjectMedical Sciences
dc.subjectMedicine and Health Sciences
dc.subjectMolecular Genetics
dc.subjectOncology
dc.subjectMIEN1
dc.subjectAnnexin A2
dc.subjectbreast cancer
dc.subjectmigration
dc.subjectinvasion
dc.subjectactin
dc.titleMIEN1 Drives Breast Cancer Invasion by Regulating Cytoskeletal-Focal Adhesions Dynamics
dc.typeDissertation
dc.type.materialtext
thesis.degree.departmentGraduate School of Biomedical Sciences
thesis.degree.disciplineCancer Biology
thesis.degree.grantorUniversity of North Texas Health Science Center at Fort Worth
thesis.degree.nameDoctor of Philosophy

Files

Original bundle

Now showing 1 - 1 of 1
Loading...
Thumbnail Image
Name:
2015_05_gsbs_Kpetemey_Marilyne_dissertation.pdf
Size:
22.07 MB
Format:
Adobe Portable Document Format