Antisense masking of an hnRNP A1/A2 intronic splicing silencer corrects SMN2 splicing in transgenic mice - PubMed (original) (raw)

Antisense masking of an hnRNP A1/A2 intronic splicing silencer corrects SMN2 splicing in transgenic mice

Yimin Hua et al. Am J Hum Genet. 2008 Apr.

Abstract

Survival of motor neuron 2, centromeric (SMN2) is a gene that modifies the severity of spinal muscular atrophy (SMA), a motor-neuron disease that is the leading genetic cause of infant mortality. Increasing inclusion of SMN2 exon 7, which is predominantly skipped, holds promise to treat or possibly cure SMA; one practical strategy is the disruption of splicing silencers that impair exon 7 recognition. By using an antisense oligonucleotide (ASO)-tiling method, we systematically screened the proximal intronic regions flanking exon 7 and identified two intronic splicing silencers (ISSs): one in intron 6 and a recently described one in intron 7. We analyzed the intron 7 ISS by mutagenesis, coupled with splicing assays, RNA-affinity chromatography, and protein overexpression, and found two tandem hnRNP A1/A2 motifs within the ISS that are responsible for its inhibitory character. Mutations in these two motifs, or ASOs that block them, promote very efficient exon 7 inclusion. We screened 31 ASOs in this region and selected two optimal ones to test in human SMN2 transgenic mice. Both ASOs strongly increased hSMN2 exon 7 inclusion in the liver and kidney of the transgenic animals. Our results show that the high-resolution ASO-tiling approach can identify cis-elements that modulate splicing positively or negatively. Most importantly, our results highlight the therapeutic potential of some of these ASOs in the context of SMA.

PubMed Disclaimer

Figures

Figure 1

Figure 1

Schematic Representation of the Binding Sites for the 20 MOE ASOs Used in the Initial ASO Walk along the Two Flanking Intronic Regions of Exon 7 (A) ASO walk at the end of intron 6. (B) ASO walk at the beginning of intron 7. The position of complementarity of each ASO is indicated by a horizontal line above the sequence.

Figure 2

Figure 2

Analysis of the 20 MOE ASOs by Splicing In Vitro and in Cells The diagrams on the right indicate the mobilities of the various RNA species. The percentage of exon 7 inclusion in each lane was calculated as described in Material and Methods and is indicated below each phosphorimage diagram. For the two in vivo splicing assays, each ASO at a concentration of 10 μM and 2 μg of pBabe-Puro was cotransfected with or without pEGFP-SMN2 into HEK293 cells. Two days after transfection, cells were collected for total RNA extraction, and RT-PCR was performed for the analysis of SMN2 pre-mRNA splicing patterns. (A) Each ASO at a concentration of 200 nM was tested by in vitro splicing with an SMN2 minigene substrate. ASO 00–00 was used as a negative control, and SMN1 was used as a positive control. The radiolabeled RNAs were analyzed by 8% denaturing PAGE. (B) The 20 ASOs were cotransfected with a pEGFP-SMN2 minigene, and RT-PCR products were analyzed by 8% native PAGE. (C) The effects of the 20 ASOs were analyzed with transcripts from the endogenous SMN2 gene in HEK293 cells. RT-PCR products were digested with DdeI so that SMN1 could be distinguished from SMN2 by 6% native PAGE. FL indicates full-length, and Δ7 indicates exon 7 deleted mRNA.

Figure 3

Figure 3

Effects of Mutations in and around the Intron 7 ISS on SMN2 Exon 7 Inclusion (A and C) WT and mutant intron 7 sequences. Mutations are shaded, and deletions are indicated by dashes. (B and D) WT pCI-SMN2 and mutant minigene plasmids (5 μg) were electroporated into HEK293 cells. Total RNA was collected two days after transfection and analyzed by radioactive RT-PCR. The radiolabeled PCR products were analyzed by 8% native PAGE and detected and quantitated with a phosphorimager.

Figure 4

Figure 4

Analysis of Proteins Bound to the Intron 7 ISS by RNA-Affinity Chromatography (A) Four RNA oligonucleotides corresponding to the WT and three mutant sequences shown were used for RNA-affinity chromatography. (B) Agarose beads covalently linked to the RNAs shown in (A) were incubated with HeLa cell nuclear extract under splicing conditions, and the beads were washed three times at the indicated salt concentrations. Bound proteins were eluted with SDS and analyzed by SDS-PAGE with Coomassie-Blue staining. The migration of size markers and hnRNP A1 and A2 are indicated. (C) Western-blotting analysis of the eluted proteins with monoclonal antibodies that recognize only hnRNP A2 and B1 (top), hnRNP A1 (middle), and all the hnRNP A/B family proteins (bottom); HeLa cell nuclear extract (NE) was also analyzed for the determination of the relative signals of the various hnRNP A/B family proteins in the starting material (bottom left).

Figure 5

Figure 5

Effects of hnRNP A1 or A2 Overexpression on Exon 7 Inclusion in SMN1 Minigene Transcripts HEK293 cells were transfected with 5 μg of the indicated WT or mutant pCI-SMN1 plasmids (described in Figures 3A and 3C). The indicated amounts of pCGT7-A1 (A) or pCGT7-A2 (B) expressing N-terminal T7-tagged hnRNP A1 or A2 proteins were cotransfected with the SMN1 reporters. Two days after transfection, total RNA was collected and radioactive RT-PCR was performed to measure the extent of exon 7 inclusion by 8% native PAGE and phosphorimage analysis. The expression of tagged hnRNP A1 or A2 was verified by western blotting with monoclonal antibody against the T7 tag. The histograms on the right show the corresponding quantitation from three independent experiments. Error bars represent the standard deviation.

Figure 6

Figure 6

Effect of Improving the hnRNP A1/A2 Motifs in the Intron 7 ISS on Exon 7 Splicing (A) Sequences of the WT intron 7 ISS and mutants with improved hnRNP A1/A2 motifs. The hnRNP A1/A2 motifs are underlined. Single mutations are shaded. (B and C) RT-PCR analysis showing the effects of improving the hnRNP A1/A2 motifs in either the motif 1 or motif 2 region. Mutants were tested in both SMN2 and SMN1 minigene contexts. Five micrograms of WT pCI-SMN2, pCI-SMN1, or each mutant plasmid was transfected into HEK293 cells by electroporation. Total RNA was extracted 2 days later and analyzed by radioactive RT-PCR and then 8% native PAGE and phosphorimage quantitation. (D) Quantitative data of mutagenesis analysis, including previous experiments (Figure 2), are presented as a scatter plot. The percentage of exon 7 inclusion was plotted against hnRNP A1 scores based on a PWM with background correction for the base composition of the winner pool. Data points for mutants in the motif 1 and motif 2 regions are shown as open circles and solid squares, respectively. Least-squares lines are shown for each data set (dashed line for motif 1 with R2 = 0.75, and solid line for motif 2 with R2 = 0.61).

Figure 7

Figure 7

Schematic Diagram of the In Vivo Effects of All Tested Intronic ASOs Horizontal bars represent ASOs with stimulatory effects (green), inhibitory effects (red), or neutral effects (blue). The thicker the bars, the stronger the effects. (A) ASOs targeting the 3′ region of intron 6. (B)ASOs targeting the 5′ region of intron 7. ∗ indicates the four 18-mer ASOs (08–25, 09–26, 10–27, and 11–28) that displayed the strongest stimulatory effects. # indicates the best 15-mer ASO (09–23) and the best 18-mer ASO (10–27) that were tested in hSMN2 transgenic mice (Figure 8).

Figure 8

Figure 8

Effects of ASOs 09–23 and 10–27 in hSMN2 Transgenic Mice ASOs 09–23, 10–27, control ASO 00–00, or saline was delivered intravenously via the tail vein, twice a week, at 25 mg/kg. Each ASO or saline was administered to eight mice, and tissues and organs including liver (A), kidney (B), thigh muscles (C) and spinal cord (D) were harvested after 1, 2, 3, or 4 weeks of treatment (two mice each). Total RNA was extracted from tissues with Trizol reagent, and RT-PCR was carried out with a set of h_SMN2_-specific primers. Radiolabeled PCR products were analyzed by 8% native PAGE and phosphorimaging. The histograms on the right show the corresponding quantitation. Error bars show standard deviations.

Similar articles

Cited by

References

    1. Hastings M.L., Krainer A.R. Pre-mRNA splicing in the new millennium. Curr. Opin. Cell Biol. 2001;13:302–309. - PubMed
    1. Brow D.A. Allosteric cascade of spliceosome activation. Annu. Rev. Genet. 2002;36:333–360. - PubMed
    1. Jurica M.S., Moore M.J. Pre-mRNA splicing: Awash in a sea of proteins. Mol. Cell. 2003;12:5–14. - PubMed
    1. Cartegni L., Chew S.L., Krainer A.R. Listening to silence and understanding nonsense: Exonic mutations that affect splicing. Nat. Rev. Genet. 2002;3:285–298. - PubMed
    1. Mayeda A., Krainer A.R. Regulation of alternative pre-mRNA splicing by hnRNP A1 and splicing factor SF2. Cell. 1992;68:365–375. - PubMed

Publication types

MeSH terms

Substances

LinkOut - more resources