142 research outputs found

    On variant discovery in genomes of fungal plant pathogens

    Get PDF
    Comparative genome analyses of eukaryotic pathogens including fungi and oomycetes have revealed extensive variability in genome composition and structure. The genomes of individuals from the same population can exhibit different numbers of chromosomes and different organization of chromosomal segments, defining so-called accessory compartments that have been shown to be crucial to pathogenicity in plant-infecting fungi. This high level of structural variation confers a methodological challenge for population genomic analyses. Variant discovery from population sequencing data is typically achieved using established pipelines based on the mapping of short reads to a reference genome. These pipelines have been developed, and extensively used, for eukaryote genomes of both plants and animals, to retrieve single nucleotide polymorphisms and short insertions and deletions. However, they do not permit the inference of large-scale genomic structural variation, as this task typically requires the alignment of complete genome sequences. Here, we compare traditional variant discovery approaches to a pipeline based on de novo genome assembly of short read data followed by whole genome alignment, using simulated data sets with properties mimicking that of fungal pathogen genomes. We show that the latter approach exhibits levels of performance comparable to that of read-mapping based methodologies, when used on sequence data with sufficient coverage. We argue that this approach further allows additional types of genomic diversity to be explored, in particular as long-read third-generation sequencing technologies are becoming increasingly available to generate population genomic data

    Comparison of Read Mapping and Variant Calling Tools for the Analysis of Plant NGS Data

    Get PDF
    Schilbert H, Rempel A, Pucker B. Comparison of Read Mapping and Variant Calling Tools for the Analysis of Plant NGS Data. Plants. 2020;9(4): 439.High-throughput sequencing technologies have rapidly developed during the past years and have become an essential tool in plant sciences. However, the analysis of genomic data remains challenging and relies mostly on the performance of automatic pipelines. Frequently applied pipelines involve the alignment of sequence reads against a reference sequence and the identification of sequence variants. Since most benchmarking studies of bioinformatics tools for this purpose have been conducted on human datasets, there is a lack of benchmarking studies in plant sciences. In this study, we evaluated the performance of 50 different variant calling pipelines, including five read mappers and ten variant callers, on six real plant datasets of the model organism Arabidopsis thaliana. Sets of variants were evaluated based on various parameters including sensitivity and specificity. We found that all investigated tools are suitable for analysis of NGS data in plant research. When looking at different performance metrics, BWA-MEM and Novoalign were the best mappers and GATK returned the best results in the variant calling ste

    Computational validation of clonal and subclonal copy number alterations from bulk tumor sequencing using CNAqc

    Get PDF
    Copy number alterations (CNAs) are among the most important genetic events in cancer, but their detection from sequencing data is challenging because of unknown sample purity, tumor ploidy, and general intra-tumor heterogeneity. Here, we present CNAqc, an evolution-inspired method to perform the computational validation of clonal and subclonal CNAs detected from bulk DNA sequencing. CNAqc is validated using single-cell data and simulations, is applied to over 4000 TCGA and PCAWG samples, and is incorporated into the validation process for the clinically accredited bioinformatics pipeline at Genomics England. CNAqc is designed to support automated quality control procedures for tumor somatic data validation

    A verified genomic reference sample for assessing performance of cancer panels detecting small variants of low allele frequency

    Get PDF
    BackgroundOncopanel genomic testing, which identifies important somatic variants, is increasingly common in medical practice and especially in clinical trials. Currently, there is a paucity of reliable genomic reference samples having a suitably large number of pre-identified variants for properly assessing oncopanel assay analytical quality and performance. The FDA-led Sequencing and Quality Control Phase 2 (SEQC2) consortium analyze ten diverse cancer cell lines individually and their pool, termed Sample A, to develop a reference sample with suitably large numbers of coding positions with known (variant) positives and negatives for properly evaluating oncopanel analytical performance.ResultsIn reference Sample A, we identify more than 40,000 variants down to 1% allele frequency with more than 25,000 variants having less than 20% allele frequency with 1653 variants in COSMIC-related genes. This is 5-100x more than existing commercially available samples. We also identify an unprecedented number of negative positions in coding regions, allowing statistical rigor in assessing limit-of-detection, sensitivity, and precision. Over 300 loci are randomly selected and independently verified via droplet digital PCR with 100% concordance. Agilent normal reference Sample B can be admixed with Sample A to create new samples with a similar number of known variants at much lower allele frequency than what exists in Sample A natively, including known variants having allele frequency of 0.02%, a range suitable for assessing liquid biopsy panels.ConclusionThese new reference samples and their admixtures provide superior capability for performing oncopanel quality control, analytical accuracy, and validation for small to large oncopanels and liquid biopsy assays.Peer reviewe

    Methods for Epigenetic Analyses from Long-Read Sequencing Data

    Get PDF
    Epigenetics, particularly the study of DNA methylation, is a cornerstone field for our understanding of human development and disease. DNA methylation has been included in the "hallmarks of cancer" due to its important function as a biomarker and its contribution to carcinogenesis and cancer cell plasticity. Long-read sequencing technologies, such as the Oxford Nanopore Technologies platform, have evolved the study of structural variations, while at the same time allowing direct measurement of DNA methylation on the same reads. With this, new avenues of analysis have opened up, such as long-range allele-specific methylation analysis, methylation analysis on structural variations, or relating nearby epigenetic modalities on the same read to another. Basecalling and methylation calling of Nanopore reads is a computationally expensive task which requires complex machine learning architectures. Read-level methylation calls require different approaches to data management and analysis than ones developed for methylation frequencies measured from short-read technologies or array data. The 2-dimensional nature of read and genome associated DNA methylation calls, including methylation caller uncertainties, are much more storage costly than 1-dimensional methylation frequencies. Methods for storage, retrieval, and analysis of such data therefore require careful consideration. Downstream analysis tasks, such as methylation segmentation or differential methylation calling, have the potential of benefiting from read information and allow uncertainty propagation. These avenues had not been considered in existing tools. In my work, I explored the potential of long-read DNA methylation analysis and tackled some of the challenges of data management and downstream analysis using state of the art software architecture and machine learning methods. I defined a storage standard for reference anchored and read assigned DNA methylation calls, including methylation calling uncertainties and read annotations such as haplotype or sample information. This storage container is defined as a schema for the hierarchical data format version 5, includes an index for rapid access to genomic coordinates, and is optimized for parallel computing with even load balancing. It further includes a python API for creation, modification, and data access, including convenience functions for the extraction of important quality statistics via a command line interface. Furthermore, I developed software solutions for the segmentation and differential methylation testing of DNA methylation calls from Nanopore sequencing. This implementation takes advantage of the performance benefits provided by my high performance storage container. It includes a Bayesian methylome segmentation algorithm which allows for the consensus instance segmentation of multiple sample and/or haplotype assigned DNA methylation profiles, while considering methylation calling uncertainties. Based on this segmentation, the software can then perform differential methylation testing and provides a large number of options for statistical testing and multiple testing correction. I benchmarked all tools on both simulated and publicly available real data, and show the performance benefits compared to previously existing and concurrently developed solutions. Next, I applied the methods to a cancer study on a chromothriptic cancer sample from a patient with Sonic Hedgehog Medulloblastoma. I here report regulatory genomic regions differentially methylated before and after treatment, allele-specific methylation in the tumor, as well as methylation on chromothriptic structures. Finally, I developed specialized methylation callers for the combined DNA methylation profiling of CpG, GpC, and context-free adenine methylation. These callers can be used to measure chromatin accessibility in a NOMe-seq like setup, showing the potential of long-read sequencing for the profiling of transcription factor co-binding. In conclusion, this thesis presents and subsequently benchmarks new algorithmic and infrastructural solutions for the analysis of DNA methylation data from long-read sequencing

    Comprehensive identification and characterisation of germline structural variation within the Iberian population

    Get PDF
    [eng] One of the central aims of biology and biomedicine has been the characterisation and understanding of genetic variation across humans, to answer important evolutionary questions and to explain phenotypic variability concerning the diseases. Understanding genetic variability, is key to study this relationship (through imputation and GWASs) and to translate the results into improved clinical protocols. Different initiatives have emerged around the world to systematically characterise the genetic variability of specific human populations from whole-genome sequences, usually by selecting geographical regions. Examples such as 1000 Genomes (1000G)1, GoNL2, HRC, UK10K3 or Estonian population4, have already identified and characterised millions of genetic variants across different populations. In combination with imputation analysis, these sequenced-based projects allow increasing the statistical power and resolution of Genome-Wide Association Studies (GWAS), identifying and discovering new disease-associated variants5. Additionally, genetic variability among population groups is associated with geographic ancestry and can affect the disease risk or treatment efficacy differently6,7. For this reason, population- specific reference panels are necessary to characterise their genetic diversity and to assess its effect on human phenotypes, improving GWAS studies, as one of the cornerstones of precision medicine7. Existing genetic variability panels include Single Nucleotide Variants (SNVs) and indels (<50bp) but are limited in large Structural Variants (SV) (≄50bp). Technical and methodological limitations hindered the discovery of SVs using Next-generation Sequencing (NGS) technologies, as it produced False-Discovery Rates between 9-89% and recall 10-70%, depending on the SV type and size8. On average, the genomic variation between two human genomes is around 0.1%, but this difference increases to 1.5% with SVs8. The SVs also affect 3-10 times more nucleotides than SNVs9 (4M SNVs per genome10), showing their potential effect on human phenotypes. For this reason, including a complete catalogue of SVs in reference panels will increase the power in GWAS studies and provide opportunities to find new disease-associated variants. To overcome these limitations, in this thesis, we have generated the first genome-wide Iberian haplotype reference panel, mainly focused on Structural Variants, using 785 samples whole-genome sequenced (WGS) at high coverage (30X) from the GCAT-Genomics for life project. We designed a complete strategy, including an extensive benchmarking of multiple variant calling programs and by building specific Logistic Regression Models (LRM) for SV types, as well as phasing strategies to come up with a high quality and comprehensive genetic variability panel. This strategy was benchmarked using different controlled sets of variants, showing high precision and recall values across all variant types and sizes. The application of this strategy to our GCAT whole-genome samples resulted in the identification of 35,431,441 genetic variants, classified as 30,325,064 SNPs, 5,017,19 small indels (< 50bp), and 89,178 larger SV (≄ 50bp). The latter group was further subclassified into 33,244 deletions, 6,269 duplications, 12,782 insertions, 10,115 inversions, 18,779 transposons and 7,989 translocations, covering all ranges of frequencies and sizes. Besides, 60% of the discovered SVs were not catalogued in any repository, thus increasing the insights of SV in humans. Additionally, 52.44% of common and 71.63% of low-frequency SVs were not included in any haplotype reference panel. Thus, new SVs could be used in GWAS, adding more value to the Iberian-GCAT catalogue. The prediction of the functional impact of the SVs shows that these variants might have a central role in several diseases. Of all SVs included in the Iberian-GCAT catalogue, 46% overlapped in genes (both protein-coding genes and non-protein-coding genes), highlighting their potential impact on human traits. Besides, 92.7% of protein-coding genes were located outside low-complexity (repeated) genomic regions, expecting short-reads from NGS to capture the most interpretable SVs in humans11. Moreover, 32.93% of SVs affected protein-coding genes with a predicted loss of function intolerance (pLI) effect, further supporting the potential implication of these variants on complex diseases and therefore enabling a better explanation of missing heritability. Importantly, taking advantage of high coverage (30X), we accurately determine the genotypes of SVs, enabling to phase together with SNVs and indels, and increasing the SV phasing accuracy, in contrast to 1000G and GoNL. Besides, high coverage allowed to use Phasing Informative Reads (PIRs), increasing the phasing performance. The overall strategy enables the community to expand and improve the imputation possibilities within GWAS. The Iberian-GCAT haplotype reference panel created in this thesis, imputes accurately common SVs, with near ~100% of agreement with sequencing results. Although the Iberian- GCAT haplotype reference panel can be used in all populations from different continental groups, due to closer ancestries, the imputation performance is high in European and Latin American populations, reflected in the amount of low-frequency (1% ≀ MAF MAF) variants imputed at high info scores. These results demonstrated the versatility of our resource, increasing their performance in closer ancestries. In general, we observed that when the allele frequency decreases, the imputation accuracy drops too, highlighting the necessity to include more samples in reference panels, to impute low-frequency and rare variants efficiently, which normally are expected to have more functional impact on diseases. Finally, we compared the imputation possibilities of the 1000G and GoNL reference panels, with our Iberian-GCAT reference panel. We observed that the Iberian-GCAT reference panel outperformed the imputation of high-quality SVs by 2.7 and 1.6-fold compared to 1000G and GoNL, respectively. Also, the overall imputation quality is higher, showing the value of this new resource in GWAS as it includes more SVs than previous reference panels. The combination of different reference panels will improve the resolution and statistical power of GWAS, thus increasing the chances to find more risk variants in complex diseases, and ultimately, translated this insight to precision medicine

    Somatic Copy Number Alteration detection and Copy Number signature analysis in High-Grade Serous Ovarian Cancer

    Get PDF
    Somatic copy number alterations (sCNAs) are a type of genomic variation that affects the dosage of DNA sequences promoting tumorigenesis such as in High grade serous ovarian cancer. Their complexity prevents the unravelling of the mechanisms generating them and the molecular stratification of the patients. Here we propose the implementation of a highly sensitive pipeline for their detection based on structural variant calling and a signature analysis for the extraction of recurrent patterns. The precise calling allowed the recovery of small segments missed from the previous pipeline and in turn the clear distinction of patients with Homologous Recombination Deficiency (HRD), which resulted extremely segmented. Although the signature analysis, based on COSMIC copy number signatures, did not provide results consistent with the clinical data, the de novo signature extraction provided 15 new signatures able to proficiently explain the dataset. Two of them were positively associated with HRD, possibly representing a test for the identification of the HRD phenotype. Further insights on these signatures may provide the discovery of their etiology and give the possibility to shed light on association with single nucleotide variations.Somatic copy number alterations (sCNAs) are a type of genomic variation that affects the dosage of DNA sequences promoting tumorigenesis such as in High grade serous ovarian cancer. Their complexity prevents the unravelling of the mechanisms generating them and the molecular stratification of the patients. Here we propose the implementation of a highly sensitive pipeline for their detection based on structural variant calling and a signature analysis for the extraction of recurrent patterns. The precise calling allowed the recovery of small segments missed from the previous pipeline and in turn the clear distinction of patients with Homologous Recombination Deficiency (HRD), which resulted extremely segmented. Although the signature analysis, based on COSMIC copy number signatures, did not provide results consistent with the clinical data, the de novo signature extraction provided 15 new signatures able to proficiently explain the dataset. Two of them were positively associated with HRD, possibly representing a test for the identification of the HRD phenotype. Further insights on these signatures may provide the discovery of their etiology and give the possibility to shed light on association with single nucleotide variations

    Data analysis methods for copy number discovery and interpretation

    Get PDF
    Copy number variation (CNV) is an important type of genetic variation that can give rise to a wide variety of phenotypic traits. Differences in copy number are thought to play major roles in processes that involve dosage sensitive genes, providing beneficial, deleterious or neutral modifications to individual phenotypes. Copy number analysis has long been a standard in clinical cytogenetic laboratories. Gene deletions and duplications can often be linked with genetic Syndromes such as: the 7q11.23 deletion of Williams-­‐Bueren Syndrome, the 22q11 deletion of DiGeorge syndrome and the 17q11.2 duplication of Potocki-­‐Lupski syndrome. Interestingly, copy number based genomic disorders often display reciprocal deletion / duplication syndromes, with the latter frequently exhibiting milder symptoms. Moreover, the study of chromosomal imbalances plays a key role in cancer research. The datasets used for the development of analysis methods during this project are generated as part of the cutting-­‐edge translational project, Deciphering Developmental Disorders (DDD). This project, the DDD, is the first of its kind and will directly apply state of the art technologies, in the form of ultra-­‐high resolution microarray and next generation sequencing (NGS), to real-­‐time genetic clinical practice. It is collaboration between the Wellcome Trust Sanger Institute (WTSI) and the National Health Service (NHS) involving the 24 regional genetic services across the UK and Ireland. Although the application of DNA microarrays for the detection of CNVs is well established, individual change point detection algorithms often display variable performances. The definition of an optimal set of parameters for achieving a certain level of performance is rarely straightforward, especially where data qualities vary ... [cont.]
    • 

    corecore