Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Dev Cell ; 36(1): 50-62, 2016 Jan 11.
Article in English | MEDLINE | ID: mdl-26766442

ABSTRACT

Control of cell-division orientation is integral to epithelial morphogenesis and asymmetric cell division. Proper spatiotemporal localization of the evolutionarily conserved Gαi-LGN-NuMA protein complex is critical for mitotic spindle orientation, but how this is achieved remains unclear. Here we identify Suppressor APC domain containing 2 (SAPCD2) as a previously unreported LGN-interacting protein. We show that SAPCD2 is essential to instruct planar mitotic spindle orientation in both epithelial cell cultures and mouse retinal progenitor cells in vivo. Loss of SAPCD2 randomizes spindle orientation, which in turn disrupts cyst morphogenesis in three-dimensional cultures, and triples the number of terminal asymmetric cell divisions in the developing retina. Mechanistically, we show that SAPCD2 negatively regulates the localization of LGN at the cell cortex, likely by competing with NuMA for its binding. These results uncover SAPCD2 as a key regulator of the ternary complex controlling spindle orientation during morphogenesis and asymmetric cell divisions.


Subject(s)
Antigens, Nuclear/metabolism , Cell Polarity/physiology , Mitosis/physiology , Nuclear Matrix-Associated Proteins/metabolism , Nuclear Proteins/metabolism , Spindle Apparatus/metabolism , Animals , Cell Cycle/genetics , Cell Cycle Proteins , Cell Polarity/genetics , Humans , Mice , Morphogenesis/physiology , Nuclear Proteins/genetics , Protein Binding
2.
J Cell Biol ; 199(6): 951-67, 2012 Dec 10.
Article in English | MEDLINE | ID: mdl-23209302

ABSTRACT

The small GTPase Rap1 regulates inside-out integrin activation and thereby influences cell adhesion, migration, and polarity. Several Rap1 effectors have been described to mediate the cellular effects of Rap1 in a context-dependent manner. Radil is emerging as an important Rap effector implicated in cell spreading and migration, but the molecular mechanisms underlying its functions are unclear. We report here that the kinesin KIF14 associates with the PDZ domain of Radil and negatively regulates Rap1-mediated inside-out integrin activation by tethering Radil on microtubules. The depletion of KIF14 led to increased cell spreading, altered focal adhesion dynamics, and inhibition of cell migration and invasion. We also show that Radil is important for breast cancer cell proliferation and for metastasis in mice. Our findings provide evidence that the concurrent up-regulation of Rap1 activity and increased KIF14 levels in several cancers is needed to reach optimal levels of Rap1-Radil signaling, integrin activation, and cell-matrix adhesiveness required for tumor progression.


Subject(s)
Adenocarcinoma/metabolism , Breast Neoplasms/metabolism , Carrier Proteins/metabolism , Kinesins/metabolism , Oncogene Proteins/metabolism , rap1 GTP-Binding Proteins/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/secondary , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carrier Proteins/genetics , Cell Line, Tumor , Cell Proliferation , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mice , Mice, Inbred NOD , Mice, SCID , Signal Transduction , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...