2021
DOI: 10.1093/nar/gkab078
|View full text |Cite
|
Sign up to set email alerts
|

Binding of phosphorothioate oligonucleotides with RNase H1 can cause conformational changes in the protein and alter the interactions of RNase H1 with other proteins

Abstract: We recently found that toxic PS-ASOs can cause P54nrb and PSF nucleolar mislocalization in an RNase H1-dependent manner. To better understand the underlying mechanisms of these observations, here we utilize different biochemical approaches to demonstrate that PS-ASO binding can alter the conformations of the bound proteins, as illustrated using recombinant RNase H1, P54nrb, PSF proteins and various isolated domains. While, in general, binding of PS-ASOs or ASO/RNA duplexes stabilizes the conformations of these… Show more

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
2
2
1

Citation Types

2
14
0

Year Published

2021
2021
2024
2024

Publication Types

Select...
8

Relationship

3
5

Authors

Journals

citations
Cited by 11 publications
(16 citation statements)
references
References 36 publications
2
14
0
Order By: Relevance
“…The reported interaction between NONO and 2′-F-PS-ASOs leads to a reduction in the nuclear abundance of NONO, likely due to targeted protein degradation ( 33 ). While the binding of the 2′-F-PS-ASOs to the NONO homodimer described in this study did not appear to compromise the integrity of the dimer, it is possible that the large-scale nuclear mRNP aggregates formed with ASO treatment elicit a defensive response to mitigate the formation of pathological aggregates, or that PS-ASO binding may affect the NONO interaction with other partner proteins ( 77 ). Ultimately, further investigation is required to explore what effect 2′-F-ASO binding to NONO containing dimers has within a cellular context.…”
Section: Discussionmentioning
confidence: 76%
“…The reported interaction between NONO and 2′-F-PS-ASOs leads to a reduction in the nuclear abundance of NONO, likely due to targeted protein degradation ( 33 ). While the binding of the 2′-F-PS-ASOs to the NONO homodimer described in this study did not appear to compromise the integrity of the dimer, it is possible that the large-scale nuclear mRNP aggregates formed with ASO treatment elicit a defensive response to mitigate the formation of pathological aggregates, or that PS-ASO binding may affect the NONO interaction with other partner proteins ( 77 ). Ultimately, further investigation is required to explore what effect 2′-F-ASO binding to NONO containing dimers has within a cellular context.…”
Section: Discussionmentioning
confidence: 76%
“…83 While 2 0 modifications have been shown previously not to affect RNase H1 binding affinity to the duplex, addition of the methyl group at the 2 0 position of the ribose would disrupt this series of interactions and require RNase H1 to traverse the hybrid to establish the necessary contacts for cleavage. 91 This alone does not suggest that overall cleavage should be reduced if RNase H1 is able to compensate by cleaving at alternative positions within the hybrid; however, in the case of the PS-ASO 558807 and its size-matched target RNA from the Cxcl12 gene, we do observe reduced cleavage over time. This could be because RNase H1 requires a minimum length of 7 to 10 RNA:DNA hybridized nucleotides to bind with its hybrid binding domain and cleave the RNA downstream.…”
Section: Discussionmentioning
confidence: 57%
“…In the case of RNase competent, the degradation of RNA is catalyzed by the RNase H enzyme RNASEH1 in presence of the RNA-DNA duplex formed by the binding of the DNA-based ONs with their specific mRNA transcripts. Thus, the target gene expression is silenced ( Figure 1 A) [ 8 , 9 ]. A central DNA-based “gap” surrounded by flanking regions, constituted by chemically modified RNA promoting target binding, constitutes the recently proposed RNase H-competent ASOs [ 10 ].…”
Section: On Therapeuticsmentioning
confidence: 99%