scholarly journals CRISPR-Cas9 cytidine and adenosine base editing of splice-sites mediates highly-efficient disruption of proteins in primary cells

Author(s):  
Mitchell G. Kluesner ◽  
Walker S. Lahr ◽  
Cara-Lin Lonetree ◽  
Branden A. Smeester ◽  
Patricia N. Claudio-Vázquez ◽  
...  

ABSTRACTBase editors allow for precise nucleotide editing without the need for genotoxic double-stranded breaks. Prior work has used base editors to knockout genes by introducing premature stop codons or by disrupting conserved splice-sites, but no direct comparison exists between these methods. Additionally, while base editor mediated disruption of splice sites has been used to shift the functional isoform pool, its utility for gene knockout requires further validation. To address these needs, we developed the program SpliceR (z.umn.edu/spliceR) to design cytidine-deaminase base editor (CBE) and adenosine-deaminase base editor (ABE) splice-site targeting guides. We compared the splice-site targeting and premature stop codon introduction in a knockout screen against the TCR-CD3 immune synapse in primary human T-cells. Our data suggests that 1) the CBE, BE4 is more reliable than the ABE, ABE7.10 for splice-site targeting knockout and 2) for both CBEs and ABEs, splice-donor targeting is the most reliable approach for base editing induced knockout.

1996 ◽  
Vol 75 (06) ◽  
pp. 870-876 ◽  
Author(s):  
José Manuel Soria ◽  
Lutz-Peter Berg ◽  
Jordi Fontcuberta ◽  
Vijay V Kakkar ◽  
Xavier Estivill ◽  
...  

SummaryNonsense mutations, deletions and splice site mutations are a common cause of type I protein C deficiency. Either directly or indirectly by altering the reading frame, these' lesions generate or may generate premature stop codons and could therefore be expected to result in premature termination of translation. In this study, the possibility that such mutations could instead exert their pathological effects at an earlier stage in the expression pathway, through “allelic exclusion” at the RNA level, was investigated. Protein C (PROC) mRNA was analysed in seven Spanish type I protein C deficient patients heterozygous for two nonsense mutations, a 7bp deletion, a 2bp insertion and three splice site mutations. Ectopic RNA transcripts from patient and control lymphocytes were analysed by RT-PCR and direct sequencing of amplified PROC cDNA fragments. The nonsense mutations and the deletion were absent from the cDNAs indicating that only mRNA derived from the normal allele had been expressed. Similarly for the splice site mutations, only normal PROC cDNAs were obtained. In one case, exclusion of the mutated allele could be confirmed by polymorphism analysis. In contrast to these six mutations, the 2 bp insertion was not associated with loss of mRNA from the mutated allele. In this case, cDNA analysis revealed the absence of 19 bases from the PROC mRNA consistent with the generation and utilization of a cryptic splice site 3’ to the site of mutation, which would result in a frameshift and a premature stop codon. It is concluded that allelic exclusion is a common causative mechanism in those cases of type I protein C deficiency which result from mutations that introduce premature stop codons


2021 ◽  
Vol 19 (1) ◽  
Author(s):  
Abhishek Kumar ◽  
Nagarajan Paramasivam ◽  
Obul Reddy Bandapalli ◽  
Matthias Schlesner ◽  
Tianhui Chen ◽  
...  

Abstract Background The most frequently identified strong cancer predisposition mutations for colorectal cancer (CRC) are those in the mismatch repair (MMR) genes in Lynch syndrome. Laboratory diagnostics include testing tumors for immunohistochemical staining (IHC) of the Lynch syndrome-associated DNA MMR proteins and/or for microsatellite instability (MSI) followed by sequencing or other techniques, such as denaturing high performance liquid chromatography (DHPLC), to identify the mutation. Methods In an ongoing project focusing on finding Mendelian cancer syndromes we applied whole-exome/whole-genome sequencing (WES/WGS) to 19 CRC families. Results Three families were identified with a pathogenic/likely pathogenic germline variant in a MMR gene that had previously tested negative in DHPLC gene variant screening. All families had a history of CRC in several family members across multiple generations. Tumor analysis showed loss of the MMR protein IHC staining corresponding to the mutated genes, as well as MSI. In family A, a structural variant, a duplication of exons 4 to 13, was identified in MLH1. The duplication was predicted to lead to a frameshift at amino acid 520 and a premature stop codon at amino acid 539. In family B, a 1 base pair deletion was found in MLH1, resulting in a frameshift and a stop codon at amino acid 491. In family C, we identified a splice site variant in MSH2, which was predicted to lead loss of a splice donor site. Conclusions We identified altogether three pathogenic/likely pathogenic variants in the MMR genes in three of the 19 sequenced families. The MLH1 variants, a duplication of exons 4 to 13 and a frameshift variant, were novel, based on the InSiGHT and ClinVar databases; the MSH2 splice site variant was reported by a single submitter in ClinVar. As a variant class, duplications have rarely been reported in the MMR gene literature, particularly those covering several exons.


Hemoglobin ◽  
2012 ◽  
Vol 36 (3) ◽  
pp. 244-252 ◽  
Author(s):  
Talal Qadah ◽  
Jill Finlayson ◽  
Christopher Newbound ◽  
Nicole Pell ◽  
Michelle Pascoe ◽  
...  

2020 ◽  
Vol 21 (1) ◽  
Author(s):  
Arthur Jacob ◽  
Jennifer Pasquier ◽  
Raphael Carapito ◽  
Frédéric Auradé ◽  
Anne Molitor ◽  
...  

Abstract Background Mandibulofacial dysostosis with microcephaly (MFDM) is a rare autosomal dominant genetic disease characterized by intellectual and growth retardations, as well as major microcephaly, induced by missense and splice site variants or microdeletions in the EFTUD2 gene. Case presentation Here, we investigate the case of a young girl with symptoms of MFDM and a normal karyotype. Whole-exome sequencing of the family was performed to identify genetic alterations responsible for this phenotype. We identified a de novo synonymous variant in the EFTUD2 gene. We demonstrated that this synonymous variant disrupts the donor splice-site in intron 9 resulting in the skipping of exon 9 and a frameshift that leads to a premature stop codon. Conclusions We present the first case of MFDM caused by a synonymous variant disrupting the donor splice site, leading to exon skipping.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2555-2555
Author(s):  
Jacob Grinfeld ◽  
Juan M Alonso-Dominguez ◽  
Mary Alikian ◽  
Mikel Valgañón ◽  
Georgios Nteliopoulos ◽  
...  

Abstract Abstract 2555 Background: The prognosis for patients with chronic myeloid leukaemia (CML) has vastly improved through the use of tyrosine kinase inhibitors (TKIs), such as imatinib. However, a proportion of patients will not respond, or will lose their initial response, and the biological mechanisms underlying this heterogeneity are poorly understood. Human organic cation transporter-1 (hOCT-1 or SLC22A1), the main transporter for imatinib, has been proposed as one determinant. This study set out to assess the prognostic value of hOCT-1 expression and the presence of polymorphisms in the hOCT-1 gene. Methods: hOCT-1 mRNA levels in 153 diagnostic whole blood samples from two different patient cohorts (one from a trial population, and the other from patients treated only at our institution) were measured by RT-qPCR (normalised against two control genes). hOCT-1 exon 7 DNA (and cDNA transcripts) were sequenced in 156 patients, and four cells lines, to identify insertions/deletions or single nucleotide polymorphisms (SNPs). Fragment length analysis using gene-scanner technology was also used to identify patients with insertions/deletions and to correlate genotypes with cDNA transcript lengths. Time to each endpoint (remission/imatinib failure) was compared according to the level of expression for each gene, or according to genotype, using Kaplan-Meier analyses and the log-rank test. Results and Conclusions: No significant differences in outcomes were found when comparing patients with high or low hOCT-1 expression (whether defined using the median or other cut-offs). The 408V>M (g.1222G>A) SNP in hOCT-1 exon 7 was found in 102/156 patients (22 homozygotes and 80 heterozygotes) and was associated in all cases with an eight base-pair insertion at the exon-intron boundary (rs113569197). This insertion was found to create an alternative splice site, leading to the transcription of an additional RNA/cDNA transcript in these patients, the sequence of which contains a premature stop codon soon after the splice site. Additionally, M420del was found in 52 patients (three homozygotes and 49 heterozygotes) and was not found in alleles containing the eight base pair insertion. The six possible combinations of these three alleles (N=no insertion/deletion, 8+=8bp insertion and 3−=3bp deletion) were found to give rise to five possible combinations of RNA transcript lengths, since patients with 8+8+ produce identical transcript lengths to those with 8+N (i.e. the normal transcript and one with an additional 8bp and premature stop). In the trial cohort (n=109), significant differences in time to 10% (p=0.01), 1% (p=0.0002) and 0.1% (p=0.0003) molecular responses (by the international scale) and time to imatinib failure (p=0.02) were seen when patients with 8+8+/8+N were compared to those with the remaining four genotypes (NN/N3−/3−8+/3−3−). However, this association was not replicated in the internal cohort, which was smaller (n=47) and more heterogeneous in terms of baseline characteristics and management. These findings may explain discrepancies in the results of previous studies that have examined the association between hOCT-1 expression and outcome, since a number used primer/probe sets that would be affected by the presence/absence of these polymorphisms. Our results suggest that while hOCT-1 expression is not a determinant of response, alterations in splice sites or amino acid sequence due to insertions/deletions may be. Further work is required to clarify the impact of these polymorphisms on hOCT-1 protein levels, function and responses to imatinib. Disclosures: White: Novartis Oncology: Honoraria, Research Funding; BMS: Research Funding; CSL: Research Funding. Marin:Novartis: Research Funding; BMS: Research Funding. Apperley:Novartis, Bristol Myers-Squibb, and ARIAD: Honoraria, Research Funding. Goldman:Novartis, Bristol Myers-Squibb, and Amgen: Honoraria.


2016 ◽  
Vol 54 (9) ◽  
pp. 1435-1440 ◽  
Author(s):  
Zhihong Wang ◽  
Yanhong Lin ◽  
Liping Qiu ◽  
Dezhu Zheng ◽  
Aizhen Yan ◽  
...  

AbstractBackground:Duchenne muscular dystrophy (DMD) is typically caused by disrupting the reading frame of the dystrophin gene: approximately 70%–80% of mutational events are represented by deletions or duplications of one or more exons in the dystrophin gene, and the remaining cases by subtle mutations, including point mutations, small indels, small inversions, and complex small rearrangements. The dystrophin gene is the largest known gene with one of the highest known rates of new mutations.Methods:Deletions and duplications were detected in theDMDgene of the proband by using multiple ligation-dependent probe amplification (MLPA). Targeted next-generation sequencing (NGS) was used in the subtle mutation detection, followed by Sanger sequencing confirmation. The effect of the mutation on the splicing of theDMDgene was assessed by bioinformatics prediction and hybrid minigene splicing assay (HMSA).Results:Neither duplication nor deletion was found in theDMDgene of the proband. While a novel splice site mutation c.6762+1G>C was identified in the proband by NGS and Sanger sequencing, and his mother was heterozygous at the same site. Bioinformatics predicted that the 5′ donor splice site of intron 46 disappeared because of the mutation, which would lead to aberrant splicing and introduce premature stop codon. The HMSA results were in agreement with the prediction.Conclusions:The novel splice site mutation caused DMD in the proband by aberrant splicing. We suggested that combined applications of MLPA, NGS, HMSA and bioinformatics are comprehensive and effective methods for diagnosis and aberrant splicing study of DMD.


2020 ◽  
Vol 11 (5-6) ◽  
pp. 264-270
Author(s):  
Jamie Fitzgerald ◽  
Cori Feist ◽  
Paula Dietz ◽  
Stephen Moore ◽  
Donald Basel

We report a novel intronic variant in the <i>MTM1</i> gene in 4 males in a family with severe X-linked myotubular myopathy. The A&#x3e;G variant in deep intronic space activates a cryptic 5′ donor splice site resulting in the inclusion of a 48-bp pseudoexon into the mature <i>MTM1</i> mRNA. The variant is present in all affected males, absent in unaffected males, and heterozygous in the mother of the affected males. The included intronic sequence contains a premature stop codon, and experiments using a translational inhibitor indicate that the mutant mRNAs undergo nonsense-mediated decay. We conclude that affected males produce no, or low, levels of <i>MTM1</i> mRNA likely leading to a significant reduction of myotubularin-1 protein resulting in the severe neonatal myopathy present in this family. The study highlights the need to consider noncoding variants in genomic screening in families with X-linked myotubular myopathy.


2006 ◽  
Vol 96 (12) ◽  
pp. 711-716 ◽  
Author(s):  
Lisa Gallinaro ◽  
Francesca Sartorello ◽  
Elena Pontara ◽  
Maria Cattini ◽  
Antonella Bertomoro ◽  
...  

SummaryWe describe the complex picture associated with a mutated splice junction in intron 13 of von Willebrand factor (VWF) gene. The proband, characterized by a marked decrease in plasma and platelet VWF and near normal multimer organization, was classified as recessive type 1 von Willebrand disease (VWD). Genetic analysis demonstrated that he was homozygous for the 1534–3C>A mutation in the consensus sequence of the acceptor splicing site of intron 13 of the VWF gene. Platelet mRNA analysis documented three VWF transcripts: a wild type generated by the correct recognition of the mutated splice site, a smaller transcript not containing exon 14, and a longer one that, in addition to exons 13 and 14, included a 62bp fragment corresponding to the end of intron 13. The small transcript derives from the skipping of exon 14, the long one from the activation of a cryptic splice site in intron 13; both show a premature stop codon inVWF propeptide, so the probandVWF derives entirely from the correct splice site recognition. Combined incomplete exon skipping and cryptic splice site activation are first recognized in VWD. Since the 1534–3C>A mutation does not abolish the normal processing of mRNA, it is unlikely to be found in type 3VWD. This mutation therefore appears to be peculiar to type 1 VWD.


2018 ◽  
Vol 19 (8) ◽  
pp. 2259 ◽  
Author(s):  
Noeline Nadarajah ◽  
Dörte Schulte ◽  
Vivienne McConnell ◽  
Silvia Martin-Almedina ◽  
Christina Karapouliou ◽  
...  

Lymphedema is characterized by chronic swelling of any body part caused by malfunctioning or obstruction in the lymphatic system. Primary lymphedema is often considered genetic in origin. VEGFC, which is a gene encoding the ligand for the vascular endothelial growth factor receptor 3 (VEGFR3/FLT4) and important for lymph vessel development during lymphangiogenesis, has been associated with a specific subtype of primary lymphedema. Through Sanger sequencing of a proband with bilateral congenital pedal edema resembling Milroy disease, we identified a novel mutation (NM_005429.2; c.361+5G>A) in VEGFC. The mutation induced skipping of exon 2 of VEGFC resulting in a frameshift and the introduction of a premature stop codon (p.Ala50ValfsTer18). The mutation leads to a loss of the entire VEGF-homology domain and the C-terminus. Expression of this Vegfc variant in the zebrafish floorplate showed that the splice-site variant significantly reduces the biological activity of the protein. Our findings confirm that the splice-site variant, c.361+5G>A, causes the primary lymphedema phenotype in the proband. We examine the mutations and clinical phenotypes of the previously reported cases to review the current knowledge in this area.


Sign in / Sign up

Export Citation Format

Share Document