2023
DOI: 10.1101/2023.02.20.529064
|View full text |Cite
Preprint
|
Sign up to set email alerts
|

Enterovirus Evolution Reveals the Mechanism of an RNA-Targeted Antiviral and Determinants of Viral Replication

Abstract: Selective pressures on positive-strand RNA viruses provide opportunities to establish target site specificity and mechanisms of action of antivirals. Here, Enterovirus-A71 revertant viruses with resistant mutations in the SLII IRES domain (SLIIresist) were selected at low doses of the antiviral DMA-135. The EV-A71 revertant viruses were resistant to DMA-135 at concentrations that robustly inhibit replication of wild-type virus. EV-A71 IRES structures harboring the suppressor mutations induced efficient express… Show more

Help me understand this report
View published versions

Search citation statements

Order By: Relevance

Paper Sections

Select...
1
1
1
1

Citation Types

0
6
0

Year Published

2023
2023
2024
2024

Publication Types

Select...
2
1
1

Relationship

1
3

Authors

Journals

citations
Cited by 4 publications
(6 citation statements)
references
References 31 publications
0
6
0
Order By: Relevance
“…In parallel with the research on new targets, a focus must be as well the research of new molecules specifically acting on viral IRES in order not to risk targeting human IRES, which are involved in the translation of genes involved in stress and apoptosis [90], processes often ongoing in infected cells. Despite part of the compounds listed in Table 2 showing some aspecific activity, the results of prunin [7] and DMA-135 [13,74] are encouraging since a specific viral RNA targeted mechanism of action was identified thanks to the selection of resistance. These results prove that achieving specific viral inhibition by targeting the viral IRES is possible and the activity can be maintained up to in vivo [7].…”
Section: Internal Ribosomal Entry Sitementioning
confidence: 99%
See 2 more Smart Citations
“…In parallel with the research on new targets, a focus must be as well the research of new molecules specifically acting on viral IRES in order not to risk targeting human IRES, which are involved in the translation of genes involved in stress and apoptosis [90], processes often ongoing in infected cells. Despite part of the compounds listed in Table 2 showing some aspecific activity, the results of prunin [7] and DMA-135 [13,74] are encouraging since a specific viral RNA targeted mechanism of action was identified thanks to the selection of resistance. These results prove that achieving specific viral inhibition by targeting the viral IRES is possible and the activity can be maintained up to in vivo [7].…”
Section: Internal Ribosomal Entry Sitementioning
confidence: 99%
“…The selection of resistance in vitro could reveal if the site targeted can be easily mutated, therefore hampering the further development of the molecule as an antiviral. However, the eventual selection of resistance is an important verification of the binding site that can be then re-engineered in dual luciferase assays or modeled to understand the change of the binding pocket of the molecule, as was previously done for inhibitors of the RNA of enterovirus 71 [7,13]. Importantly if the resistance mutation leads to the identification or validation of the binding site, it can also open the possibility of modifying the small molecule with a medicinal chemistry approach that could ameliorate the inhibition profile.…”
Section: Validation With Wt Virusmentioning
confidence: 99%
See 1 more Smart Citation
“…HnRNP A1 binds to a phylogenetically conserved bulge loop to change the structure of SLII, which in turn stimulates IRES activity. Mutations or deletions to the bulge loop abrogate hnRNP A1 binding, significantly attenuate IRES translation and inhibit EV-A1 replication by ~5 log 10 units [ 15 , 53 , 56 ]. Thus, the recruitment of hnRNP A1 to the EV-A71 5′-UTR is essential for efficient viral replication.…”
Section: Cellular Ires Trans-acting Factors (Itafs) That Regulate Ev-...mentioning
confidence: 99%
“…Further, this region has been successfully targeted by a small molecule antiviral DMA-135 (2-log and 5-log reduction of viral titers at 0.5 and 50 µM of DMA-135, respectively; IC 50 of 7.54 ± 0.0024 μM) where its mechanism of action is to allosterically increase the binding affinity of AUF1 to SLII, thus shifting the SLII regulatory axis towards translation repression [ 64 ]. Notably, EV-A71 can evolve drug resistance to DMA-135 by changing nucleotides that form part of the AUF1 binding site, while not perturbing the binding activity of hnRNP A1 [ 56 ]. These collective observations suggest that other small molecules with the capacity to selectively perturb specific 5′-UTR-ITAF interactions await discovery.…”
Section: Future Perspectivesmentioning
confidence: 99%