LAUSR.org creates dashboard-style pages of related content for over 1.5 million academic articles. Sign Up to like articles & get recommendations!

Anti-Zika virus RNAi in neural progenitor cells

Photo by cdc from unsplash

Zika virus (ZIKV), a flavivirus that is primarily transmitted by mosquitoes, can cause devastating microcephaly and other congenital syndromes in infants born to infected pregnant women. In a recent paper… Click to show full abstract

Zika virus (ZIKV), a flavivirus that is primarily transmitted by mosquitoes, can cause devastating microcephaly and other congenital syndromes in infants born to infected pregnant women. In a recent paper in Cell Research, Xu et al. present data demonstrating that (i) ZIKV infection induces RNA interference (RNAi)-mediated antiviral defense in human neural progenitor cells and (ii) treatment with enoxacin, a broad-spectrum antibiotic with known RNAi-enhancing activity, prevents ZIKV infection and microcephalic defect in brain organoids. Zika virus (ZIKV) has recently emerged as a global public health threat. Due to the explosive epidemics and teratogenic potential, the World Health Organization declared ZIKV to be a Public Health Emergency of International Concern in 2016. Besides ZIKV, many other flaviviruses are also significant human pathogens, including dengue, yellow fever, West Nile, Japanese encephalitis, and tickborne encephalitis viruses. Compared with other flaviviruses, ZIKV is unique in causing congenital syndrome in fetuses of infected pregnant women, including microcephaly, craniofacial disproportion, ocular abnormalities, spasticity, and miscarriage. Adults infected with ZIKV may develop Guillain–Barré syndrome, an autoimmune disorder caused by the immune system damaging the peripheral nervous system. The U.S. Centers for Disease Control and Prevention estimated one in seven infants born to mothers infected during pregnancy may develop birth defects. Upon maternal-to-fetal transmission in pregnant women, ZIKV preferentially infects human neural progenitor cells (hNPCs) in the fetal brain. The infection impairs hNPC proliferation and differentiation, triggers apoptosis of progeny cells, and debilitates brain development. Thus, it is critical to define the immune response when hNPCs are infected by ZIKV. Such knowledge is critical to understand ZIKV-mediated congenital syndrome as well as to develop novel countermeasures. All flaviviruses produce double-stranded RNA (dsRNA) as a replication intermediate during infection. The dsRNA serves as a pathogen-associated molecular pattern (PAMP) to elicit interferon (IFN)-mediated antiviral defense in mammalian host. Although RNA interference (RNAi) has been well established as an evolutionarily conserved innate immune response in fungi, plants, and invertebrates, its role in antiviral defense in mammalians remains to be fully established. To achieve this goal, Xu and colleagues have now provided three sets of compelling evidence supporting that ZIKV infection induces RNAi-mediated antiviral response in hNPCs. First, Xu et al. demonstrated an abundant production of virusderived small interfering RNAs (vsiRNAs) in ZIKV-infected hNPCs (Fig. 1a). vsiRNAs were measured by deep sequencing and validated by northern blot. Approximately 63% of the virusderived small RNA (vsRNA) reads were 21–23 nt in length, with predominant 22-nt vsRNAs representing both positiveand negative-strand RNAs. However, once hNPCs had been differentiated into neurons, they lost the ability to produce vsiRNAs after ZIKV infection. This result suggests a switch-off of the vsiRNA biogenesis pathway during the hNPC-to-neuron differentiation. The conclusion is further supported by the results that no vsiRNA was detected in ZIKV-infected human rhabdomyoma RD cells or astrocytes. Altogether, the data indicate that hNPCs represent a unique cell population that has a functional RNAi machinery to process viral dsRNAs to vsiRNAs. These vsiRNAs are then loaded onto the RNA-induced silencing complexes (RISCs) to initiate the cleavage of viral RNA in infected hNPCs (Fig. 1a). Second, Xu et al. showed that the production of vsiRNAs is dependent on the authentic RNAi pathway in ZIKV-infected hNPCs (Fig. 1a). Knockdown or knockout of Dicer or AGO2, the two essential components of RNAi pathway, significantly enhanced ZIKV replication in hNPCs. In addition, ectopic expression of the Nodamura virus B2 protein, a known viral suppressor of RNAi (VSR), increased ZIKV replication in hNPCs. These results demonstrate that (i) an intact RNAi pathway is required for the production of vsiRNAs in ZIKV-infected hNPCs and (ii) the vsiRNAs are functional in inhibiting ZIKV infection. Third, Xu et al. evinced a therapeutic approach using an RNAi enhancer to treat ZIKV infection in hNPCs and brain organoids (Fig. 1b). Enoxacin is a clinically approved antibiotic with known RNAi-enhancing activity, possibly through modulating PIWIL3 (piwi like RNA-mediated gene silencing 3, an Argonaute protein of the PIWI subfamily). In hNPCs, treatment with enoxacin suppressed ZIKV infection with IC50 of 52 μM and CC50 of 176 μM. However, the compound failed to inhibit ZIKV in Dicerknockout hNPCs, indicating that the observed antiviral activity is dependent on the RNAi pathway. In brain organoids, enoxacin inhibited ZIKV infection of hNPCs in the ventricular zone (VZ) and subventricular zone (SVZ), rescued hNPC proliferation, and maintained a normal brain development. These promising results warrant further in vivo efficacy testing of enoxacin in mouse or non-human primate microcephaly models. The findings by Xu et al. have opened new directions for future research. First, IFN pathway has been well established as the major innate immunity in mammalians. However, recent studies indicate that stem cells are refractory to IFN, but intrinsically express a subset of IFN-stimulated genes (ISGs) for antiviral defense. The discovery of functional vsiRNAs in ZIKV-infected hNPCs suggests that, together with distinct ISGs in stem cells, RNAi may serve as another arm of antiviral defense. It remains to be determined (i) whether the results observed in hNPCs could be generalized to stem cells of other tissue types and (ii) whether other viruses

Keywords: hnpcs; infection; zikv infection; zikv; brain; virus

Journal Title: Cell Research
Year Published: 2019

Link to full text (if available)


Share on Social Media:                               Sign Up to like & get
recommendations!

Related content

More Information              News              Social Media              Video              Recommended



                Click one of the above tabs to view related content.