scholarly journals The regulation of focal adhesion complex formation and salivary gland epithelial cell organization by nanofibrous PLGA scaffolds

Biomaterials ◽  
2012 ◽  
Vol 33 (11) ◽  
pp. 3175-3186 ◽  
Author(s):  
Sharon J. Sequeira ◽  
David A. Soscia ◽  
Basak Oztan ◽  
Aaron P. Mosier ◽  
Riffard Jean-Gilles ◽  
...  
2005 ◽  
Vol 206 (2) ◽  
pp. 510-517 ◽  
Author(s):  
Zhen Wang ◽  
Kenton D. Fong ◽  
Toan-Thang Phan ◽  
Ivor J. Lim ◽  
Michael T. Longaker ◽  
...  

1999 ◽  
Vol 83 (12) ◽  
pp. 38-43 ◽  
Author(s):  
Toshio Nagai ◽  
Martin Laser ◽  
Catalin F Baicu ◽  
Michael R Zile ◽  
George Cooper ◽  
...  

2010 ◽  
Vol 298 (4) ◽  
pp. C831-C846 ◽  
Author(s):  
Sharon Israeli ◽  
Kurt Amsler ◽  
Nadezhda Zheleznova ◽  
Patricia D. Wilson

Integrin-associated focal adhesion complex formation and turnover plays an essential role in directing interactions between epithelial cells and the extracellular matrix during organogenesis, leading to appropriate cell spreading, cell-matrix adhesion, and migration. Autosomal recessive polycystic kidney disease (ARPKD) is associated with loss of function of PKHD1-encoded protein fibrocystin-1 and is characterized by cystic dilation of renal collecting tubules (CT) in utero and loss of renal function in patients if they survive the perinatal period. Normal polycystin-1 (PC-1)/focal adhesion complex function is required for control of CT diameter during renal development, and abnormalities in these complexes have been demonstrated in human PC-1 mutant cystic cells. To determine whether loss of fibrocystin-1 was associated with focal adhesion abnormalities, ARPKD cells or normal age-matched human fetal (HF)CT cells in which fibrocystin-1 had been decreased by 85% by small interfering RNA inhibition were compared with normal HFCT. Accelerated attachment and spreading on collagen matrix and decreased motility of fibrocystin-1-deficient cells were associated with longer paxillin-containing focal adhesions, more complex actin-cytoskeletal rearrangements, and increased levels of total β1-integrin, c-Src, and paxillin. Immunoblot analysis of adhesive cells using site-specific phospho-antibodies demonstrated ARPKD-associated loss of activation of focal adhesion kinase (FAK) by phosphorylation at its autophosphorylation site (Y397); accelerated FAK inhibition by phosphorylation at Y407, S843, and S910; as well as increased activation of c-Src at pY418. Paxillin coimmunoprecipitation analyses suggested that fibrocystin-1 was a component of the normal focal adhesion complex and that actin and fibrocystin-1 were lost from ARPKD complexes.


2020 ◽  
Vol 117 (16) ◽  
pp. 9064-9073
Author(s):  
David de Semir ◽  
Vladimir Bezrookove ◽  
Mehdi Nosrati ◽  
Kara R. Scanlon ◽  
Eric Singer ◽  
...  

The invasive behavior of glioblastoma is essential to its aggressive potential. Here, we show that pleckstrin homology domain interacting protein (PHIP), acting through effects on the force transduction layer of the focal adhesion complex, drives glioblastoma motility and invasion. Immunofluorescence analysis localized PHIP to the leading edge of glioblastoma cells, together with several focal adhesion proteins: vinculin (VCL), talin 1 (TLN1), integrin beta 1 (ITGB1), as well as phosphorylated forms of paxillin (pPXN) and focal adhesion kinase (pFAK). Confocal microscopy specifically localized PHIP to the force transduction layer, together with TLN1 and VCL. Immunoprecipitation revealed a physical interaction between PHIP and VCL. Targeted suppression of PHIP resulted in significant down-regulation of these focal adhesion proteins, along with zyxin (ZYX), and produced profoundly disorganized stress fibers. Live-cell imaging of glioblastoma cells overexpressing a ZYX-GFP construct demonstrated a role for PHIP in regulating focal adhesion dynamics. PHIP silencing significantly suppressed the migratory and invasive capacity of glioblastoma cells, partially restored following TLN1 or ZYX cDNA overexpression. PHIP knockdown produced substantial suppression of tumor growth upon intracranial implantation, as well as significantly reduced microvessel density and secreted VEGF levels. PHIP copy number was elevated in the classical glioblastoma subtype and correlated with elevated EGFR levels. These results demonstrate PHIP’s role in regulating the actin cytoskeleton, focal adhesion dynamics, and tumor cell motility, and identify PHIP as a key driver of glioblastoma migration and invasion.


Sign in / Sign up

Export Citation Format

Share Document