Abstract

Influenza A virus (IAV) causes a respiratory infection that affects millions of people of different age groups and can lead to acute respiratory distress syndrome. Currently, host genes, receptors, and other cellular components critical for IAV replication are actively studied. One of the most convenient and accessible genome-editing tools to facilitate these studies is the CRISPR/Cas9 system. This tool allows for regulating the expression of both viral and host cell genes to enhance or impair viral entry and replication. This review considers the effect of the genome editing system on specific target genes in cells (human and chicken) in terms of subsequent changes in the influenza virus life cycle and the efficiency of virus particle production.

Highlights

  • Laboratory of Vector Vaccines, Smorodintsev Research Institute of Influenza, Ministry of Health of the Russian Federation, 197376 Saint Petersburg, Russia

  • Lee and colleagues performed siRNA-mediated gene silencing of cMDA5, cTLR3, and cTLR7 as well as clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9-mediated individual and double knockout of cMDA5 and cTLR3 in chicken embryonic fibroblast DF-1 cells to determine their roles in the IFN-mediated innate immunity in response to avian influenza virus infection

  • These results suggested that cTLR7 was neither involved in IFN-β signaling in response to avian influenza virus (AIV) in chicken compared to cMDA5 and cTLR3 nor played a significant role in sensing RNA ligands

Read more

Summary

Introduction

Influenza A virus (IAV) causes acute respiratory infections in humans and remains a continuous and severe threat to public health. Unlike RNAi or ASOs methods, the CRISPR/Cas system has higher specificity and fewer off-target effects [32] It helps to increase the understanding of the interaction between the virus and the host cell in terms of both regulatory factors and RNA modifications. Cas13-assisted restriction of viral expression and readout (CARVER) platform to detect and cleave the single-stranded RNA viruses such as lymphocytic choriomeningitis virus (LCMV); influenza A virus (IAV); and vesicular stomatitis virus (VSV) This technology can be used clinically to rapidly measure wild-type viral RNA levels and detect specific viral mutations, but it is limited by the toxicity of single-stranded RNA cleavage in eukaryotic cells [29]. Abbott et al [33] de of 14 veloped a prophylactic antiviral CRISPR in human cells (PAC-MAN) strategy based on the CRISPR/Cas system, which can degrade SARS-CoV2 RNA fragments and live influenza A virus. RNA marked in red is viral mRNA; RNA marked in blue is viral negative-sense RNA

Genes of the Innate Immune Response
OAS Family
Sialic Acids
Genes Responsible for Viral Penetration into the Nucleus
Splicing Regulating Genes
Genes of RNA Modification
Findings
Conclusions
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call