Skip to main content
Top
Published in: Molecular Brain 1/2021

Open Access 01-12-2021 | Rabies | Methodology

Development of a rabies virus-based retrograde tracer with high trans-monosynaptic efficiency by reshuffling glycoprotein

Authors: Fan Jia, Li Li, Haizhou Liu, Pei Lv, Xiangwei Shi, Yang Wu, Chen Ling, Fuqiang Xu

Published in: Molecular Brain | Issue 1/2021

Login to get access

Abstract

Rabies 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.
Appendix
Available only for authorised users
Literature
1.
2.
go back to reference Wickersham IR, et al. Monosynaptic restriction of transsynaptic tracing from single, genetically targeted neurons. Neuron. 2007;53(5):639–47.CrossRef Wickersham IR, et al. Monosynaptic restriction of transsynaptic tracing from single, genetically targeted neurons. Neuron. 2007;53(5):639–47.CrossRef
3.
go back to reference Wickersham IR, et al. Retrograde neuronal tracing with a deletion-mutant rabies virus. Nat Methods. 2007;4(1):47–9.CrossRef Wickersham IR, et al. Retrograde neuronal tracing with a deletion-mutant rabies virus. Nat Methods. 2007;4(1):47–9.CrossRef
4.
go back to reference Reardon TR, et al. Rabies virus CVS-N2c(DeltaG) strain enhances retrograde synaptic transfer and neuronal viability. Neuron. 2016;89(4):711–24.CrossRef Reardon TR, et al. Rabies virus CVS-N2c(DeltaG) strain enhances retrograde synaptic transfer and neuronal viability. Neuron. 2016;89(4):711–24.CrossRef
5.
go back to reference Chatterjee S, et al. Nontoxic, double-deletion-mutant rabies viral vectors for retrograde targeting of projection neurons. Nat Neurosci. 2018;21(4):638–46.CrossRef Chatterjee S, et al. Nontoxic, double-deletion-mutant rabies viral vectors for retrograde targeting of projection neurons. Nat Neurosci. 2018;21(4):638–46.CrossRef
6.
go back to reference Ciabatti E, et al. Life-long genetic and functional access to neural circuits using self-inactivating rabies virus. Cell. 2017;170(2):382-392 e14.CrossRef Ciabatti E, et al. Life-long genetic and functional access to neural circuits using self-inactivating rabies virus. Cell. 2017;170(2):382-392 e14.CrossRef
7.
go back to reference Callaway EM, Luo L. Monosynaptic circuit tracing with glycoprotein-deleted rabies viruses. J Neurosci. 2015;35(24):8979–85.CrossRef Callaway EM, Luo L. Monosynaptic circuit tracing with glycoprotein-deleted rabies viruses. J Neurosci. 2015;35(24):8979–85.CrossRef
8.
go back to reference Hong S, et al. Functional analysis of various promoters in lentiviral vectors at different stages of in vitro differentiation of mouse embryonic stem cells. Mol Ther. 2007;15(9):1630–9.CrossRef Hong S, et al. Functional analysis of various promoters in lentiviral vectors at different stages of in vitro differentiation of mouse embryonic stem cells. Mol Ther. 2007;15(9):1630–9.CrossRef
9.
go back to reference Schlabach MR, et al. Synthetic design of strong promoters. Proc Natl Acad Sci U S A. 2010;107(6):2538–43.CrossRef Schlabach MR, et al. Synthetic design of strong promoters. Proc Natl Acad Sci U S A. 2010;107(6):2538–43.CrossRef
10.
go back to reference Francis DM, Page R. Strategies to optimize protein expression in E. coli. Curr Protoc Protein Sci. 2010;24:1–29 (chapter 5, unit 5). Francis DM, Page R. Strategies to optimize protein expression in E. coli. Curr Protoc Protein Sci. 2010;24:1–29 (chapter 5, unit 5).
11.
go back to reference Zhou Z, et al. Codon usage is an important determinant of gene expression levels largely through its effects on transcription. Proc Natl Acad Sci U S A. 2016;113(41):E6117–25.CrossRef Zhou Z, et al. Codon usage is an important determinant of gene expression levels largely through its effects on transcription. Proc Natl Acad Sci U S A. 2016;113(41):E6117–25.CrossRef
12.
go back to reference Srividya D, Shyam Mohan AH, Rao SN. Expression and purification of codon-optimized cre recombinase in E. coli. Protein Expr Purif. 2020;167:105546.CrossRef Srividya D, Shyam Mohan AH, Rao SN. Expression and purification of codon-optimized cre recombinase in E. coli. Protein Expr Purif. 2020;167:105546.CrossRef
13.
go back to reference Jia F, et al. Optimization of the fluorescent protein expression level based on pseudorabies virus bartha strain for neural circuit tracing. Front Neuroanat. 2019;13:63.CrossRef Jia F, et al. Optimization of the fluorescent protein expression level based on pseudorabies virus bartha strain for neural circuit tracing. Front Neuroanat. 2019;13:63.CrossRef
14.
go back to reference Miyamichi K, et al. Dissecting local circuits: parvalbumin interneurons underlie broad feedback control of olfactory bulb output. Neuron. 2013;80(5):1232–45.CrossRef Miyamichi K, et al. Dissecting local circuits: parvalbumin interneurons underlie broad feedback control of olfactory bulb output. Neuron. 2013;80(5):1232–45.CrossRef
15.
go back to reference Kim EJ, et al. Improved monosynaptic neural circuit tracing using engineered rabies virus glycoproteins. Cell Rep. 2016;15(4):692–9.CrossRef Kim EJ, et al. Improved monosynaptic neural circuit tracing using engineered rabies virus glycoproteins. Cell Rep. 2016;15(4):692–9.CrossRef
16.
go back to reference Kunec D, Osterrieder N. Codon pair bias is a direct consequence of dinucleotide bias. Cell Rep. 2016;14(1):55–67.CrossRef Kunec D, Osterrieder N. Codon pair bias is a direct consequence of dinucleotide bias. Cell Rep. 2016;14(1):55–67.CrossRef
17.
go back to reference Coleman JR, et al. Virus attenuation by genome-scale changes in codon pair bias. Science. 2008;320(5884):1784–7.CrossRef Coleman JR, et al. Virus attenuation by genome-scale changes in codon pair bias. Science. 2008;320(5884):1784–7.CrossRef
18.
go back to reference Le Nouen C, et al. Attenuation of human respiratory syncytial virus by genome-scale codon-pair deoptimization. Proc Natl Acad Sci U S A. 2014;111(36):13169–74.CrossRef Le Nouen C, et al. Attenuation of human respiratory syncytial virus by genome-scale codon-pair deoptimization. Proc Natl Acad Sci U S A. 2014;111(36):13169–74.CrossRef
19.
go back to reference Shen SH, et al. Large-scale recoding of an arbovirus genome to rebalance its insect versus mammalian preference. Proc Natl Acad Sci U S A. 2015;112(15):4749–54.CrossRef Shen SH, et al. Large-scale recoding of an arbovirus genome to rebalance its insect versus mammalian preference. Proc Natl Acad Sci U S A. 2015;112(15):4749–54.CrossRef
20.
go back to reference Ling C, et al. Selective in vivo targeting of human liver tumors by optimized AAV3 vectors in a murine xenograft model. Hum Gene Therapy. 2014;25(12):1023–34.CrossRef Ling C, et al. Selective in vivo targeting of human liver tumors by optimized AAV3 vectors in a murine xenograft model. Hum Gene Therapy. 2014;25(12):1023–34.CrossRef
21.
go back to reference Paxinos G, Franklin KBJ. The mouse brain in stereotaxic coordinates. 2nd ed. San Diego, Calif. London: Academic. 2001; xxv, 264 p Paxinos G, Franklin KBJ. The mouse brain in stereotaxic coordinates. 2nd ed. San Diego, Calif. London: Academic. 2001; xxv, 264 p
22.
go back to reference Qin JY, et al. Systematic comparison of constitutive promoters and the doxycycline-inducible promoter. PLoS ONE. 2010;5(5): e10611.CrossRef Qin JY, et al. Systematic comparison of constitutive promoters and the doxycycline-inducible promoter. PLoS ONE. 2010;5(5): e10611.CrossRef
23.
go back to reference Norrman K, et al. Quantitative comparison of constitutive promoters in human ES cells. PLoS ONE. 2010;5(8): e12413.CrossRef Norrman K, et al. Quantitative comparison of constitutive promoters in human ES cells. PLoS ONE. 2010;5(8): e12413.CrossRef
25.
go back to reference Gao L, et al. HP-PRRSV is attenuated by de-optimization of codon pair bias in its RNA-dependent RNA polymerase nsp9 gene. Virology. 2015;485:135–44.CrossRef Gao L, et al. HP-PRRSV is attenuated by de-optimization of codon pair bias in its RNA-dependent RNA polymerase nsp9 gene. Virology. 2015;485:135–44.CrossRef
26.
go back to reference Eschke K, et al. Attenuation of a very virulent Marek’s disease herpesvirus (MDV) by codon pair bias deoptimization. PLoS Pathog. 2018;14(1): e1006857.CrossRef Eschke K, et al. Attenuation of a very virulent Marek’s disease herpesvirus (MDV) by codon pair bias deoptimization. PLoS Pathog. 2018;14(1): e1006857.CrossRef
27.
go back to reference Meng J, et al. Refining the balance of attenuation and immunogenicity of respiratory syncytial virus by targeted codon deoptimization of virulence genes. MBio. 2014;5(5): e01704-14.CrossRef Meng J, et al. Refining the balance of attenuation and immunogenicity of respiratory syncytial virus by targeted codon deoptimization of virulence genes. MBio. 2014;5(5): e01704-14.CrossRef
28.
go back to reference Gaunt E, et al. Elevation of CpG frequencies in influenza A genome attenuates pathogenicity but enhances host response to infection. Elife. 2016;5: e12735.CrossRef Gaunt E, et al. Elevation of CpG frequencies in influenza A genome attenuates pathogenicity but enhances host response to infection. Elife. 2016;5: e12735.CrossRef
29.
go back to reference Burns CC, et al. Genetic inactivation of poliovirus infectivity by increasing the frequencies of CpG and UpA dinucleotides within and across synonymous capsid region codons. J Virol. 2009;83(19):9957–69.CrossRef Burns CC, et al. Genetic inactivation of poliovirus infectivity by increasing the frequencies of CpG and UpA dinucleotides within and across synonymous capsid region codons. J Virol. 2009;83(19):9957–69.CrossRef
30.
go back to reference Al-Saif M, Khabar KS. UU/UA dinucleotide frequency reduction in coding regions results in increased mRNA stability and protein expression. Mol Ther. 2012;20(5):954–9.CrossRef Al-Saif M, Khabar KS. UU/UA dinucleotide frequency reduction in coding regions results in increased mRNA stability and protein expression. Mol Ther. 2012;20(5):954–9.CrossRef
31.
go back to reference Tulloch F, et al. RNA virus attenuation by codon pair deoptimisation is an artefact of increases in CpG/UpA dinucleotide frequencies. Elife. 2014;3: e04531.CrossRef Tulloch F, et al. RNA virus attenuation by codon pair deoptimisation is an artefact of increases in CpG/UpA dinucleotide frequencies. Elife. 2014;3: e04531.CrossRef
32.
go back to reference Bajar BT, et al. Improving brightness and photostability of green and red fluorescent proteins for live cell imaging and FRET reporting. Sci Rep. 2016;6:20889.CrossRef Bajar BT, et al. Improving brightness and photostability of green and red fluorescent proteins for live cell imaging and FRET reporting. Sci Rep. 2016;6:20889.CrossRef
33.
go back to reference Osakada F, et al. New rabies virus variants for monitoring and manipulating activity and gene expression in defined neural circuits. Neuron. 2011;71(4):617–31.CrossRef Osakada F, et al. New rabies virus variants for monitoring and manipulating activity and gene expression in defined neural circuits. Neuron. 2011;71(4):617–31.CrossRef
Metadata
Title
Development of a rabies virus-based retrograde tracer with high trans-monosynaptic efficiency by reshuffling glycoprotein
Authors
Fan Jia
Li Li
Haizhou Liu
Pei Lv
Xiangwei Shi
Yang Wu
Chen Ling
Fuqiang Xu
Publication date
01-12-2021
Publisher
BioMed Central
Keyword
Rabies
Published in
Molecular Brain / Issue 1/2021
Electronic ISSN: 1756-6606
DOI
https://doi.org/10.1186/s13041-021-00821-7

Other articles of this Issue 1/2021

Molecular Brain 1/2021 Go to the issue