scholarly journals SvABA: Genome-wide detection of structural variants and indels by local assembly

2017 ◽  
Author(s):  
Jeremiah Wala ◽  
Pratiti Bandopadhayay ◽  
Noah Greenwald ◽  
Ryan O’Rourke ◽  
Ted Sharpe ◽  
...  

AbstractStructural variants (SVs), including small insertion and deletion variants (indels), are challenging to detect through standard alignment-based variant calling methods. Sequence assembly offers a powerful approach to identifying SVs, but is difficult to apply at-scale genome-wide for SV detection due to its computational complexity and the difficulty of extracting SVs from assembly contigs. We describe SvABA, an efficient and accurate method for detecting SVs from short-read sequencing data using genome-wide local assembly with low memory and computing requirements. We evaluated SvABA’s performance on the NA12878 human genome and in simulated and real cancer genomes. SvABA demonstrates superior sensitivity and specificity across a large spectrum of SVs, and substantially improved detection performance for variants in the 20-300 bp range, compared with existing methods. SvABA also identifies complex somatic rearrangements with chains of short (< 1,000 bp) templated-sequence insertions copied from distant genomic regions. We applied SvABA to 344 cancer genomes from 11 cancer types, and found that templated-sequence insertions occur in ~4% of all somatic rearrangements. Finally, we demonstrate that SvABA can identify sites of viral integration and cancer driver alterations containing medium-sized SVs.

2019 ◽  
Author(s):  
Yoav Voichek ◽  
Detlef Weigel

AbstractStructural variants and presence/absence polymorphisms are common in plant genomes, yet they are routinely overlooked in genome-wide association studies (GWAS). Here, we expand the genetic variants detected in GWAS to include major deletions, insertions, and rearrangements. We first use raw sequencing data directly to derive short sequences, k-mers, that mark a broad range of polymorphisms independently of a reference genome. We then link k-mers associated with phenotypes to specific genomic regions. Using this approach, we re-analyzed 2,000 traits measured in Arabidopsis thaliana, tomato, and maize populations. Associations identified with k-mers recapitulate those found with single-nucleotide polymorphisms (SNPs), however, with stronger statistical support. Moreover, we identified new associations with structural variants and with regions missing from reference genomes. Our results demonstrate the power of performing GWAS before linking sequence reads to specific genomic regions, which allow detection of a wider range of genetic variants responsible for phenotypic variation.


2019 ◽  
Author(s):  
Xin Zhou ◽  
Lu Zhang ◽  
Xiaodong Fang ◽  
Yichen Liu ◽  
David L. Dill ◽  
...  

AbstractHuman diploid genome assembly enables identifying maternal and paternal genetic variations. Algorithms based on 10x linked-read sequencing have been developed for de novo assembly, variant calling and haplotyping. Another linked-read technology, single tube long fragment read (stLFR), has recently provided a low-cost single tube solution that can enable long fragment data. However, no existing software is available for human diploid assembly and variant calls. We develop Aquila stLFR to adapt to the key characteristics of stLFR. Aquila stLFR assembles near perfect diploid assembled contigs, and the assembly-based variant calling shows that Aquila stLFR detects large numbers of structural variants which were not easily spanned by Illumina short-reads. Furthermore, the hybrid assembly mode Aquila hybrid allows a hybrid assembly based on both stLFR and 10x linked-reads libraries, demonstrating that these two technologies can always be complementary to each other for assembly to improve contiguity and the variants detection, regardless of assembly quality of the library itself from single sequencing technology. The overlapped structural variants (SVs) from two independent sequencing data of the same individual, and the SVs from hybrid assemblies provide us a high-confidence profile to study them.AvailabilitySource code and documentation are available on https://github.com/maiziex/Aquila_stLFR.


2018 ◽  
Author(s):  
Li Fang ◽  
Charlly Kao ◽  
Michael V Gonzalez ◽  
Fernanda A Mafra ◽  
Renata Pellegrino da Silva ◽  
...  

AbstractLinked-read sequencing provides long-range information on short-read sequencing data by barcoding reads originating from the same DNA molecule, and can improve the detection and breakpoint identification for structural variants (SVs). We present LinkedSV for SV detection on linked-read sequencing data. LinkedSV considers barcode overlapping and enriched fragment endpoints as signals to detect large SVs, while it leverages read depth, paired-end signals and local assembly to detect small SVs. Benchmarking studies demonstrates that LinkedSV outperforms existing tools, especially on exome data and on somatic SVs with low variant allele frequencies. We demonstrate clinical cases where LinkedSV identifies disease causal SVs from linked-read exome sequencing data missed by conventional exome sequencing, and show examples where LinkedSV identifies SVs missed by high-coverage long-read sequencing. In summary, LinkedSV can detect SVs missed by conventional short-read and long-read sequencing approaches, and may resolve negative cases from clinical genome/exome sequencing studies.


Author(s):  
Umair Ahsan ◽  
Qian Liu ◽  
Li Fang ◽  
Kai Wang

AbstractVariant (SNPs/indels) detection from high-throughput sequencing data remains an important yet unresolved problem. Long-read sequencing enables variant detection in difficult-to-map genomic regions that short-read sequencing cannot reliably examine (for example, only ~80% of genomic regions are marked as “high-confidence region” to have SNP/indel calls in the Genome In A Bottle project); however, the high per-base error rate poses unique challenges in variant detection. Existing methods on long-read data typically rely on analyzing pileup information from neighboring bases surrounding a candidate variant, similar to short-read variant callers, yet the benefits of much longer read length are not fully exploited. Here we present a deep neural network called NanoCaller, which detects SNPs by examining pileup information solely from other nonadjacent candidate SNPs that share the same long reads using long-range haplotype information. With called SNPs by NanoCaller, NanoCaller phases long reads and performs local realignment on two sets of phased reads to call indels by another deep neural network. Extensive evaluation on 5 human genomes (sequenced by Nanopore and PacBio long-read techniques) demonstrated that NanoCaller greatly improved performance in difficult-to-map regions, compared to other long-read variant callers. We experimentally validated 41 novel variants in difficult-to-map regions in a widely-used benchmarking genome, which cannot be reliably detected previously. We extensively evaluated the run-time characteristics and the sensitivity of parameter settings of NanoCaller to different characteristics of sequencing data. Finally, we achieved the best performance in Nanopore-based variant calling from MHC regions in the PrecisionFDA Variant Calling Challenge on Difficult-to-Map Regions by ensemble calling. In summary, by incorporating haplotype information in deep neural networks, NanoCaller facilitates the discovery of novel variants in complex genomic regions from long-read sequencing data.


2017 ◽  
Author(s):  
Rebecca Elyanow ◽  
Hsin-Ta Wu ◽  
Benjamin J. Raphael

AbstractStructural variation, including large deletions, duplications, inversions, translocations, and other rearrangements, is common in human and cancer genomes. A number of methods have been developed to identify structural variants from Illumina short-read sequencing data. However, reliable identification of structural variants remains challenging because many variants have breakpoints in repetitive regions of the genome and thus are difficult to identify with short reads. The recently developed linked-read sequencing technology from 10X Genomics combines a novel barcoding strategy with Illumina sequencing. This technology labels all reads that originate from a small number (~5-10) DNA molecules ~50Kbp in length with the same molecular barcode. These barcoded reads contain long-range sequence information that is advantageous for identification of structural variants. We present Novel Adjacency Identification with Barcoded Reads (NAIBR), an algorithm to identify structural variants in linked-read sequencing data. NAIBR predicts novel adjacencies in a individual genome resulting from structural variants using a probabilistic model that combines multiple signals in barcoded reads. We show that NAIBR outperforms several existing methods for structural variant identification – including two recent methods that also analyze linked-reads – on simulated sequencing data and 10X whole-genome sequencing data from the NA12878 human genome and the HCC1954 breast cancer cell line. Several of the novel somatic structural variants identified in HCC1954 overlap known cancer genes.


2022 ◽  
Author(s):  
Linyi Zhang ◽  
Samridhi Chaturvedi ◽  
Chris Nice ◽  
Lauren Lucas ◽  
Zachariah Gompert

Structural variants (SVs) can promote speciation by directly causing reproductive isolation or by suppressing recombination across large genomic regions. Whereas examples of each mechanism have been documented, systematic tests of the role of SVs in speciation are lacking. Here, we take advantage of long-read (Oxford nanopore) whole-genome sequencing and a hybrid zone between two Lycaeides butterfly taxa (L. melissa and Jackson Hole Lycaeides) to comprehensively evaluate genome-wide patterns of introgression for SVs and relate these patterns to hypotheses about speciation. We found >100,000 SVs segregating within or between the two hybridizing species. SVs and SNPs exhibited similar levels of genetic differentiation between species, with the exception of inversions, which were more differentiated. We detected credible variation in patterns of introgression among SV loci in the hybrid zone, with 562 of 1419 ancestry-informative SVs exhibiting genomic clines that deviating from null expectations based on genome-average ancestry. Overall, hybrids exhibited a directional shift towards Jackson Hole Lycaeides ancestry at SV loci, consistent with the hypothesis that these loci experienced more selection on average then SNP loci. Surprisingly, we found that deletions, rather than inversions, showed the highest skew towards excess introgression from Jackson Hole Lycaeides. Excess Jackson Hole Lycaeides ancestry in hybrids was also especially pronounced for Z-linked SVs and inversions containing many genes. In conclusion, our results show that SVs are ubiquitous and suggest that SVs in general, but especially deletions, might contribute disproportionately to hybrid fitness and thus (partial) reproductive isolation.


2017 ◽  
Author(s):  
Giuseppe Narzisi ◽  
André Corvelo ◽  
Kanika Arora ◽  
Ewa A. Bergmann ◽  
Minita Shah ◽  
...  

Reliable detection of somatic variations is of critical importance in cancer research. Lancet is an accurate and sensitive somatic variant caller which detects SNVs and indels by jointly analyzing reads from tumor and matched normal samples using colored DeBruijn graphs. Extensive experimental comparison on synthetic and real whole-genome sequencing datasets demonstrates that Lancet has better accuracy, especially for indel detection, than widely used somatic callers, such as MuTect, MuTect2, LoFreq, Strelka, and Strelka2. Lancet features a reliable variant scoring system which is essential for variant prioritization and detects low frequency mutations without sacrificing the sensitivity to call longer insertions and deletions empowered by the local assembly engine. In addition to genome-wide analysis, Lancet allows inspection of somatic variants in graph space, which augments the traditional read alignment visualization to help confirm a variant of interest. Lancet is available as an open-source program at https://github.com/nygenome/lancet.


2019 ◽  
Author(s):  
Sergey Aganezov ◽  
Benjamin J. Raphael

AbstractMany cancer genomes are extensively rearranged with highly aberrant chromosomal karyotypes. These genome rearrangements, or structural variants, can be detected in tumor DNA sequencing data by abnormal mapping of se-quence reads to the reference genome. However, nearly all cancer sequencing to date is of bulk tumor samples which consist of a heterogeneous mixture of normal cells and subpopulations of cancers cells, or clones, that harbor distinct somatic structural variants. We introduce a novel algorithm, Reconstructing Cancer Karyotypes (RCK), to reconstruct haplotype-specific karyotypes of one or more rearranged cancer genomes, or clones, that best explain the read alignments from a bulk tumor sample. RCK leverages specific evolutionary constraints on the somatic mutation process in cancer to reduce ambiguity in the deconvolution of admixed DNA sequence data into multiple haplotype-specific cancer karyotypes. In particular, RCK relies on generalizations of the infinite sites assumption that a genome re-arrangement is highly unlikely to occur at the same nucleotide position more than once during somatic evolution. RCK’s comprehensive model allows us to incorporate information both from short and long-read sequencing technologies and is applicable to bulk tumor samples containing a mixture of an arbitrary number of derived genomes. We compared RCK to the state-of-the-art method ReMixT on a dataset of 17 primary and metastatic prostate cancer samples. We demonstrate that ReMixT’s limited support for heterogeneity and lack of evolutionary constrains leads to reconstruction of implausible karyotypes. In contrast, RCK’s infers cancer karyotypes that better explain read alignments from bulk tumor samples and are consistent with a reasonable evolutionary model. RCK’s reconstructions of clone- and haplotype-specific karyotypes will aid further studies of the role of intra-tumor heterogeneity in cancer development and response to treatment. RCK is available at https://github.com/raphael-group/RCK.


2020 ◽  
Author(s):  
HoJoon Lee ◽  
Ahmed Shuaibi ◽  
John M. Bell ◽  
Dmitri S. Pavlichin ◽  
Hanlee P. Ji

ABSTRACTThe cancer genome sequencing has led to important discoveries such as identifying cancer gene. However, challenges remain in the analysis of cancer genome sequencing. One significant issue is that mutations identified by multiple variant callers are frequently discordant even when using the same genome sequencing data. For insertion and deletion mutations, oftentimes there is no agreement among different callers. Identifying somatic mutations involves read mapping and variant calling, a complicated process that uses many parameters and model tuning. To validate the identification of true mutations, we developed a method using k-mer sequences. First, we characterized the landscape of unique versus non-unique k-mers in the human genome. Second, we developed a software package, KmerVC, to validate the given somatic mutations from sequencing data. Our program validates the occurrence of a mutation based on statistically significant difference in frequency of k-mers with and without a mutation from matched normal and tumor sequences. Third, we tested our method on both simulated and cancer genome sequencing data. Counting k-mer involving mutations effectively validated true positive mutations including insertions and deletions across different individual samples in a reproducible manner. Thus, we demonstrated a straightforward approach for rapidly validating mutations from cancer genome sequencing data.


Sign in / Sign up

Export Citation Format

Share Document