2018
DOI: 10.1038/s41588-018-0160-6
|View full text |Cite
|
Sign up to set email alerts
|

Deep learning sequence-based ab initio prediction of variant effects on expression and disease risk

Abstract: Key challenges for human genetics, precision medicine and evolutionary biology include deciphering the regulatory code of gene expression and understanding the transcriptional effects of genome variation. However, this is extremely difficult because of the enormous scale of the noncoding mutation space. We developed a deep learning-based framework, ExPecto, that can accurately predict, ab initio from a DNA sequence, the tissue-specific transcriptional effects of mutations, including those that are rare or that… Show more

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
3
1
1

Citation Types

7
602
0
6

Year Published

2019
2019
2023
2023

Publication Types

Select...
5
2
1

Relationship

0
8

Authors

Journals

citations
Cited by 447 publications
(615 citation statements)
references
References 56 publications
7
602
0
6
Order By: Relevance
“…However, as we indicated in Figure 3, most of nonsynonymous mutations are rare variations. Therefore, a reliable way to predict potential pharmacogenetic variation is also important to improve the efficacy or avoid serious toxicity [23] . The third challenge is that a suitable strategy for precision medicine in COVID-19 treatment is still lack.…”
Section: Discussionmentioning
confidence: 99%
“…However, as we indicated in Figure 3, most of nonsynonymous mutations are rare variations. Therefore, a reliable way to predict potential pharmacogenetic variation is also important to improve the efficacy or avoid serious toxicity [23] . The third challenge is that a suitable strategy for precision medicine in COVID-19 treatment is still lack.…”
Section: Discussionmentioning
confidence: 99%
“…Disease risk variants identified by genome-wide association studies (GWAS) lie predominantly in non-coding regions of the genome 1,2,3,4,5,6,7 , motivating broad efforts to generate genome-wide maps of regulatory marks across tissues and cell types 8,9,10,11 . Recently, deep learning models trained using these genome-wide maps have shown considerable promise in predicting regulatory marks directly from DNA sequence 12,13,14,15,16,17,18 . In particular, these studies showed that variant-level deep learning annotations (based on the reference allele) attained high accuracy in predicting the underlying chromatin marks 13,14,15,16 , and that models incorporating allelic-effect deep learning annotations (absolute value of the predicted difference between reference and variant alleles) attained high accuracy in predicting disease-associated SNPs 13,14,15,16 ; additional applications of deep learning models, including analyses of signed allelic-effect annotations, are discussed in the Discussion section.…”
Section: Introductionmentioning
confidence: 99%
“…Recently, deep learning models trained using these genome-wide maps have shown considerable promise in predicting regulatory marks directly from DNA sequence 12,13,14,15,16,17,18 . In particular, these studies showed that variant-level deep learning annotations (based on the reference allele) attained high accuracy in predicting the underlying chromatin marks 13,14,15,16 , and that models incorporating allelic-effect deep learning annotations (absolute value of the predicted difference between reference and variant alleles) attained high accuracy in predicting disease-associated SNPs 13,14,15,16 ; additional applications of deep learning models, including analyses of signed allelic-effect annotations, are discussed in the Discussion section. However, it is unclear whether deep learning annotations at commonly varying SNPs contain unique information for complex disease that is not present in other annotations.…”
Section: Introductionmentioning
confidence: 99%
See 1 more Smart Citation
“…86 Massively parallel assays have also been used in the context of both splicing 88,89 and miRNA-mediated regulation, 90,91 showing, for instance, that up to 16% of splice disrupting variants are located in deep intronic regions. 88 With the development of deep learning frameworks for sequence-based predictions, 92,93 data generated by these assays, will now fuel the construction of predictive models. These models will, in turn, allow quantifying the regulatory impact of novel genetic variants on transcriptional activity, 86 alternative splicing, 94 or miRNA-mediated regulation.…”
Section: Deciphering the Regulatory Code To Predict The Effect Of Rmentioning
confidence: 99%