Antiviral RNA interference in disease vector (Asian longhorned) ticks

Autor: Yang Li, Liting Ma, Yan Xu, Chuang Gao, Yanxin Ren, Zhi Ye, Jingwen Wang, Zhengwei Zhong
Jazyk: angličtina
Rok vydání: 2021
Předmět:
Molecular biology
Physiology
viruses
Disease Vectors
Biochemistry
Mice
RNA interference
Sequencing techniques
Medical Conditions
Ticks
RNA Virus Infections
Medicine and Health Sciences
Nodaviridae
RNA
Small Interfering

Biology (General)
Gene knockdown
Mammalian Genomics
biology
Drosophila Melanogaster
Eukaryota
RNA sequencing
Genomics
Animal Models
Small interfering RNA
Body Fluids
Nucleic acids
Insects
Infectious Diseases
Blood
Genetic interference
Experimental Organism Systems
Tick-Borne Diseases
Host-Pathogen Interactions
Epigenetics
Drosophila
Haemaphysalis longicornis
Anatomy
Research Article
Nodamura virus
Sindbis virus
Arthropoda
Severe Fever with Thrombocytopenia Syndrome
QH301-705.5
Immunology
Tick
Microbiology
Virus
Model Organisms
Virology
Arachnida
parasitic diseases
Genetics
Animals
Non-coding RNA
Biology and life sciences
Ixodes
Organisms
RC581-607
biology.organism_classification
bacterial infections and mycoses
Invertebrates
Viral Replication
Gene regulation
Research and analysis methods
Species Interactions
Disease Models
Animal

Molecular biology techniques
Viral replication
Animal Genomics
Animal Studies
RNA
Parasitology
Gene expression
Sindbis Virus
Immunologic diseases. Allergy
Zoology
Entomology
Zdroj: PLoS Pathogens, Vol 17, Iss 12, p e1010119 (2021)
PLoS Pathogens
ISSN: 1553-7374
1553-7366
Popis: Disease vectors such as mosquitoes and ticks play a major role in the emergence and re-emergence of human and animal viral pathogens. Compared to mosquitoes, however, much less is known about the antiviral responses of ticks. Here we showed that Asian longhorned ticks (Haemaphysalis longicornis) produced predominantly 22-nucleotide virus-derived siRNAs (vsiRNAs) in response to severe fever with thrombocytopenia syndrome virus (SFTSV, an emerging tick-borne virus), Nodamura virus (NoV), or Sindbis virus (SINV) acquired by blood feeding. Notably, experimental acquisition of NoV and SINV by intrathoracic injection also initiated viral replication and triggered the production of vsiRNAs in H. longicornis. We demonstrated that a mutant NoV deficient in expressing its viral suppressor of RNAi (VSR) replicated to significantly lower levels than wildtype NoV in H. longicornis, but accumulated to higher levels after knockdown of the tick Dicer2-like protein identified by phylogeny comparison. Moreover, the expression of a panel of known animal VSRs in cis from the genome of SINV drastically enhanced the accumulation of the recombinant viruses. This study establishes a novel model for virus-vector-mouse experiments with longhorned ticks and provides the first in vivo evidence for an antiviral function of the RNAi response in ticks. Interestingly, comparing the accumulation levels of SINV recombinants expressing green fluorescent protein or SFTSV proteins identified the viral non-structural protein as a putative VSR. Elucidating the function of ticks’ antiviral RNAi pathway in vivo is critical to understand the virus-host interaction and the control of tick-borne viral pathogens.
Author summary Tick-borne diseases (TBDs) are the most common illnesses transmitted by ticks, and the annual number of reported TBD cases continues to increase. The Asian longhorned tick, a vector associated with at least 30 human pathogens, is native to eastern Asia and recently reached the USA as an emerging disease threat. Newly identified tick-transmitted pathogens continue to be reported, raising concerns about how TBDs occur. Interestingly, tick can harbor pathogens without being affected themselves. For viral infections, ticks have their own immune systems that protect them from infection. Meanwhile, tick-borne viruses have evolved to avoid these defenses as they establish themselves within the vector. Here, we show in detail that infecting longhorned ticks with distinct arthropod-borne RNA viruses through two approaches natural blood feeding and injection, all induce the production of vsiRNAs. Dicer2-like homolog plays a role in regulating antiviral RNAi responses as knocking down of this gene enhanced viral replication. Furthermore, we demonstrate that tick antiviral RNAi responses are inhibited through expression heterologous VSR proteins in recombinant SINV. We identify both the virus and tick factors are critical components to understanding TBDs. Importantly, our study introduces a novel, in vivo virus-vector-mouse model system for exploring TBDs in the future.
Databáze: OpenAIRE
Nepřihlášeným uživatelům se plný text nezobrazuje