scholarly journals Mouse model of ocular hypertension with retinal ganglion cell degeneration

PLoS ONE ◽  
2019 ◽  
Vol 14 (1) ◽  
pp. e0208713 ◽  
Author(s):  
Ryo Mukai ◽  
Dong Ho Park ◽  
Yoko Okunuki ◽  
Eiichi Hasegawa ◽  
Garrett Klokman ◽  
...  
2020 ◽  
Vol 11 (9) ◽  
Author(s):  
Meng-Lu Zhang ◽  
Guo-Li Zhao ◽  
Yu Hou ◽  
Shu-Min Zhong ◽  
Lin-Jie Xu ◽  
...  

Abstract Autophagy has a fundamental role in maintaining cell homeostasis. Although autophagy has been implicated in glaucomatous pathology, how it regulates retinal ganglion cell (RGC) injury is largely unknown. In the present work, we found that biphasic autophagy in RGCs occurred in a mouse model of chronic ocular hypertension (COH), accompanied by activation of Rac1, a member of the Rho family. Rac1 conditional knockout (Rac1 cKO) in RGCs attenuated RGC apoptosis, in addition to blocking the increase in the number of autophagosomes and the expression of autophagy-related proteins (Beclin1, LC3-II/I, and p62) in COH retinas. Electron micrograph and double immunostaining of LAMP1 and LC3B showed that Rac1 cKO accelerated autolysosome fusion in RGC axons of COH mice. Inhibiting the first autophagic peak with 3-methyladenine or Atg13 siRNA reduced RGC apoptosis, whereas inhibiting the second autophagic peak with 3-MA or blocking autophagic flux by chloroquine increased RGC apoptosis. Furthermore, Rac1 cKO reduced the number of autophagosomes and apoptotic RGCs induced by rapamycin injected intravitreally, which suggests that Rac1 negatively regulates mTOR activity. Moreover, Rac1 deletion decreased Bak expression and did not interfere with the interaction of Beclin1 and Bcl-2 or Bak in COH retinas. In conclusion, autophagy promotes RGC apoptosis in the early stages of glaucoma and results in autophagic cell death in later stages. Rac1 deletion alleviates RGC damage by regulating the cross talk between autophagy and apoptosis through mTOR/Beclin1-Bak. Interfering with the Rac1/mTOR signaling pathway may provide a new strategy for treating glaucoma.


2017 ◽  
Vol 8 (7) ◽  
pp. e2945-e2945 ◽  
Author(s):  
Stephanie B Syc-Mazurek ◽  
Kimberly A Fernandes ◽  
Richard T Libby

2010 ◽  
Vol 90 (1) ◽  
pp. 168-183 ◽  
Author(s):  
Manuel Salinas-Navarro ◽  
Luis Alarcón-Martínez ◽  
Francisco J. Valiente-Soriano ◽  
Manuel Jiménez-López ◽  
Sergio Mayor-Torroglosa ◽  
...  

Gene Therapy ◽  
2021 ◽  
Author(s):  
Shagana Visuvanathan ◽  
Adam N. Baker ◽  
Pamela S. Lagali ◽  
Stuart G. Coupland ◽  
Garfield Miller ◽  
...  

2016 ◽  
Vol 143 ◽  
pp. 28-38 ◽  
Author(s):  
Andrew Osborne ◽  
Marina Hopes ◽  
Phillip Wright ◽  
David C. Broadway ◽  
Julie Sanderson

2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Michal Geva ◽  
Noga Gershoni-Emek ◽  
Luana Naia ◽  
Philip Ly ◽  
Sandra Mota ◽  
...  

AbstractOptic neuropathies such as glaucoma are characterized by retinal ganglion cell (RGC) degeneration and death. The sigma-1 receptor (S1R) is an attractive target for treating optic neuropathies as it is highly expressed in RGCs, and its absence causes retinal degeneration. Activation of the S1R exerts neuroprotective effects in models of retinal degeneration. Pridopidine is a highly selective and potent S1R agonist in clinical development. We show that pridopidine exerts neuroprotection of retinal ganglion cells in two different rat models of glaucoma. Pridopidine strongly binds melanin, which is highly expressed in the retina. This feature of pridopidine has implications to its ocular distribution, bioavailability, and effective dose. Mitochondria dysfunction is a key contributor to retinal ganglion cell degeneration. Pridopidine rescues mitochondrial function via activation of the S1R, providing support for the potential mechanism driving its neuroprotective effect in retinal ganglion cells.


Sign in / Sign up

Export Citation Format

Share Document