scholarly journals A Phosphotyrosine-dependent Protein Interaction Screen Reveals a Role for Phosphorylation of Caveolin-1 on Tyrosine 14

2002 ◽  
Vol 277 (11) ◽  
pp. 8771-8774 ◽  
Author(s):  
Haiming Cao ◽  
William E. Courchesne ◽  
Cynthia Corley Mastick
2019 ◽  
Vol 14 (12) ◽  
pp. 2729-2736 ◽  
Author(s):  
Hideki Watanabe ◽  
Chuya Yoshida ◽  
Ayako Ooishi ◽  
Yasuto Nakai ◽  
Momoko Ueda ◽  
...  

2005 ◽  
Vol 17 (1) ◽  
pp. 23-35 ◽  
Author(s):  
Wei Jiang ◽  
Raffaella Sordella ◽  
Guang-Chao Chen ◽  
Shweta Hakre ◽  
Ananda L. Roy ◽  
...  

2012 ◽  
Vol 199 (3) ◽  
pp. 425-435 ◽  
Author(s):  
Bharat Joshi ◽  
Michele Bastiani ◽  
Scott S. Strugnell ◽  
Cecile Boscher ◽  
Robert G. Parton ◽  
...  

Caveolin-1 (Cav1) is an essential component of caveolae whose Src kinase-dependent phosphorylation on tyrosine 14 (Y14) is associated with regulation of focal adhesion dynamics. However, the relationship between these disparate functions remains to be elucidated. Caveola biogenesis requires expression of both Cav1 and cavin-1, but Cav1Y14 phosphorylation is dispensable. In this paper, we show that Cav1 tyrosine phosphorylation induces caveola biogenesis via actin-dependent mechanotransduction and inactivation of the Egr1 (early growth response-1) transcription factor, relieving inhibition of endogenous Cav1 and cavin-1 genes. Cav1 phosphorylation reduces Egr1 binding to Cav1 and cavin-1 promoters and stimulates their activity. In MDA-231 breast carcinoma cells that express elevated levels of Cav1 and caveolae, Egr1 regulated Cav1, and cavin-1 promoter activity was dependent on actin, Cav1, Src, and Rho-associated kinase as well as downstream protein kinase C (PKC) signaling. pCav1 is therefore a mechanotransducer that acts via PKC to relieve Egr1 transcriptional inhibition of Cav1 and cavin-1, defining a novel feedback regulatory loop to regulate caveola biogenesis.


PLoS ONE ◽  
2010 ◽  
Vol 5 (12) ◽  
pp. e15566 ◽  
Author(s):  
Masahiro Inoue ◽  
Kouichi Yasuda ◽  
Haruki Uemura ◽  
Natsumi Yasaka ◽  
Hiroshi Inoue ◽  
...  

2003 ◽  
Vol 15 (3) ◽  
pp. 289-298 ◽  
Author(s):  
Amy R. Sanguinetti ◽  
Cynthia Corley Mastick

2021 ◽  
Author(s):  
Tae-Young Ha ◽  
Yu Ree Choi ◽  
Hye Rin Noh ◽  
Seon-Heui Cha ◽  
Jae-Bong Kim ◽  
...  

Abstract Parkinson's disease (PD) is the second most prevalent neurodegenerative disease, with aging being considered the greatest risk factor for developing PD. Caveolin-1 (Cav-1) is known to participate in the aging process. Recent evidence indicates that prion-like propagation of misfolded α-synuclein (α-syn) released from neurons to neighboring neurons plays an important role in PD progression. In the present study, we demonstrated that cav-1 expression in the brain increased with age, and considerably increased in the brain of A53T α-syn transgenic mice. Cav-1 overexpression facilitated the uptake of α-syn into neurons and formation of additional Lewy body-like inclusion bodies, phosphorylation of cav-1 at tyrosine 14 was found to be crucial for this process. This study demonstrates the relationship between age and α-syn spread and will facilitate our understanding of the molecular mechanism of the cell-to-cell transmission of α-syn.


2013 ◽  
Vol 27 (S1) ◽  
Author(s):  
Suzanne E Rohrback ◽  
Hemal H Patel ◽  
Piyush M Patel ◽  
Brian P Head

2020 ◽  
Author(s):  
Tae-Young Ha ◽  
Yu Ree Choi ◽  
Hye Rin Noh ◽  
Seon-Heui Cha ◽  
Jae-Bong Kim ◽  
...  

Abstract Background: Parkinson's disease (PD) is the second most prevalent neurodegenerative disease, with aging being considered the greatest risk factor for developing PD. Caveolin-1 (Cav-1) is known to participate in the aging process. Recent evidence indicates that prion-like propagation of misfolded α-synuclein (α-syn) released from neurons to neighboring neurons plays an important role in PD progression. In the present study, we explored the association between cav-1 and cell-to-cell transmission of α-syn.Methods: Using SH-SY5Y cells and primary neurons overexpressing WT and Y14A cav-1, we investigated the effect of cav-1 expression on the uptake of α-syn using a dual chamber system. Additionally, we investigated the effect of cav-1 expression on the formation of Lewy body-like inclusions using co-culture assay and microfluidic chamber assay.Results: We demonstrated that cav-1 expression in the brain increased with age, and considerably increased in the brain of A53T α-syn transgenic mice. Cav-1 overexpression facilitated the uptake of α-syn into neurons and formation of additional Lewy body-like inclusion bodies, phosphorylation of cav-1 at tyrosine 14 was found to be crucial for this process. Conclusions: This study demonstrates the relationship between age and α-syn spread and will facilitate our understanding of the molecular mechanism of the cell-to-cell transmission of α-syn.


2015 ◽  
Vol 29 (S1) ◽  
Author(s):  
Adriana Zimnicka ◽  
Yawer Husain ◽  
Ayesha Shajahan ◽  
Peter Toth ◽  
Richard Minshall

2019 ◽  
Vol 40 (2) ◽  
pp. 420-436 ◽  
Author(s):  
Yutaro Hoshi ◽  
Yasuo Uchida ◽  
Masanori Tachikawa ◽  
Sumio Ohtsuki ◽  
Pierre-Olivier Couraud ◽  
...  

Exposure of the brain to high levels of glucocorticoids during ischemia–reperfusion induces neuronal cell death. Oxidative stress alters blood–brain barrier (BBB) function during ischemia–reperfusion, and so we hypothesized that it might impair P-glycoprotein (P-gp)-mediated efflux transport of glucocorticoids at the BBB. Therefore, the purpose of this study was to clarify the molecular mechanism of this putative decrease of P-gp-mediated efflux function. First, we established that H2O2 treatment of a human in vitro BBB model (hCMEC/D3) reduced both P-gp efflux transport activity and protein expression on the plasma membrane within 20 min. These results suggested that the rapid decrease of efflux function might be due to internalization of P-gp. Furthermore, H2O2 treatment markedly increased tyrosine-14-phosphorylated caveolin-1, which is involved in P-gp internalization. A brain perfusion study in rats showed that cortisol efflux at the BBB was markedly decreased by H2O2 administration, and inhibitors of Abl kinase and Src kinase, which phosphorylate tyrosine-14 in caveolin-1, suppressed this decrease. Overall, these findings support the idea that oxidative stress-induced activation of Abl kinase and Src kinase induces internalization of P-gp via the phosphorylation of tyrosine-14 in caveolin-1, leading to a rapid decrease of P-gp-mediated cortisol efflux at the BBB.


Sign in / Sign up

Export Citation Format

Share Document