scholarly journals New Rabies Virus Variants for Monitoring and Manipulating Activity and Gene Expression in Defined Neural Circuits

Neuron ◽  
2011 ◽  
Vol 71 (4) ◽  
pp. 617-631 ◽  
Author(s):  
Fumitaka Osakada ◽  
Takuma Mori ◽  
Ali H. Cetin ◽  
James H. Marshel ◽  
Beatriz Virgen ◽  
...  
Neuron ◽  
2012 ◽  
Vol 74 (1) ◽  
pp. 206 ◽  
Author(s):  
Fumitaka Osakada ◽  
Takuma Mori ◽  
Ali H. Cetin ◽  
James H. Marshel ◽  
Beatriz Virgen ◽  
...  

2021 ◽  
Vol 14 (1) ◽  
Author(s):  
Fan Jia ◽  
Li Li ◽  
Haizhou Liu ◽  
Pei Lv ◽  
Xiangwei Shi ◽  
...  

AbstractRabies virus (RV) is the most widely used vector for mapping neural circuits. Previous studies have shown that the RV glycoprotein can be a target to improve the retrograde transsynaptic tracing efficiency. However, the current versions still label only a small portion of all presynaptic neurons. Here, we reshuffled the oG sequence, a chimeric glycoprotein, with positive codon pair bias score (CPBS) based on bioinformatic analysis of mouse codon pair bias, generating ooG, a further optimized glycoprotein. Our experimental data reveal that the ooG has a higher expression level than the oG in vivo, which significantly increases the tracing efficiency by up to 12.6 and 62.1-fold compared to oG and B19G, respectively. The new tool can be used for labeling neural circuits Therefore, the approach reported here provides a convenient, efficient and universal strategy to improve protein expression for various application scenarios such as trans-synaptic tracing efficiency, cell engineering, and vaccine and oncolytic virus designs.


PLoS ONE ◽  
2015 ◽  
Vol 10 (5) ◽  
pp. e0128020 ◽  
Author(s):  
Sho Sato ◽  
Shinya Ohara ◽  
Ken-Ichiro Tsutsui ◽  
Toshio Iijima

2005 ◽  
Vol 15 (5) ◽  
pp. 599-606 ◽  
Author(s):  
John F Guzowski ◽  
Jerilyn A Timlin ◽  
Badri Roysam ◽  
Bruce L McNaughton ◽  
Paul F Worley ◽  
...  

2019 ◽  
Author(s):  
Rajeevkumar Raveendran Nair ◽  
Stefan Blankvoort ◽  
Maria Jose Lagartos ◽  
Cliff Kentros

SummaryUnderstanding brain function requires understanding neural circuits at the level of specificity at which they operate. While recent years have seen the development of a variety of remarkable molecular tools for the study of neural circuits, their utility is currently limited by the inability to deploy them in specific elements of native neural circuits, i.e. particular neuronal subtypes. One can obtain a degree of specificity with neuron-specific promoters, but native promoters are almost never sufficiently specific restricting this approach to transgenic animals. We recently showed that one can obtain transgenic mice with augmented anatomical specificity in targeted brain regions by identifyingcis-regulatory elements (i.e. enhancers) uniquely active in those brain regions and combining them with a heterologous promoter, an approach we call EDGE (Enhancer-Driven Gene Expression). Here we extend this strategy to the generation of viral (rAAV) vectors, showing that when combined with the right minimal promoter they largely recapitulate the specificity seen in the corresponding transgenic lines in wildtype animals, even of another species. Because active enhancers can be identified in any tissue sample, this approach promises to enable the kind of circuit-specific manipulations in any species. This should not only greatly enhance our understanding of brain function, but may one day even provide novel therapeutic avenues to correct the imbalances in neural circuits underlying many disorders of the brain.


Sign in / Sign up

Export Citation Format

Share Document