925 research outputs found

    Multiscale, multimodal analysis of tumor heterogeneity in IDH1 mutant vs wild-type diffuse gliomas.

    Get PDF
    Glioma is recognized to be a highly heterogeneous CNS malignancy, whose diverse cellular composition and cellular interactions have not been well characterized. To gain new clinical- and biological-insights into the genetically-bifurcated IDH1 mutant (mt) vs wildtype (wt) forms of glioma, we integrated data from protein, genomic and MR imaging from 20 treatment-naïve glioma cases and 16 recurrent GBM cases. Multiplexed immunofluorescence (MxIF) was used to generate single cell data for 43 protein markers representing all cancer hallmarks, Genomic sequencing (exome and RNA (normal and tumor) and magnetic resonance imaging (MRI) quantitative features (protocols were T1-post, FLAIR and ADC) from whole tumor, peritumoral edema and enhancing core vs equivalent normal region were also collected from patients. Based on MxIF analysis, 85,767 cells (glioma cases) and 56,304 cells (GBM cases) were used to generate cell-level data for 24 biomarkers. K-means clustering was used to generate 7 distinct groups of cells with divergent biomarker profiles and deconvolution was used to assign RNA data into three classes. Spatial and molecular heterogeneity metrics were generated for the cell data. All features were compared between IDH mt and IDHwt patients and were finally combined to provide a holistic/integrated comparison. Protein expression by hallmark was generally lower in the IDHmt vs wt patients. Molecular and spatial heterogeneity scores for angiogenesis and cell invasion also differed between IDHmt and wt gliomas irrespective of prior treatment and tumor grade; these differences also persisted in the MR imaging features of peritumoral edema and contrast enhancement volumes. A coherent picture of enhanced angiogenesis in IDHwt tumors was derived from multiple platforms (genomic, proteomic and imaging) and scales from individual proteins to cell clusters and heterogeneity, as well as bulk tumor RNA and imaging features. Longer overall survival for IDH1mt glioma patients may reflect mutation-driven alterations in cellular, molecular, and spatial heterogeneity which manifest in discernable radiological manifestations

    Medoidshift clustering applied to genomic bulk tumor data.

    Get PDF
    Despite the enormous medical impact of cancers and intensive study of their biology, detailed characterization of tumor growth and development remains elusive. This difficulty occurs in large part because of enormous heterogeneity in the molecular mechanisms of cancer progression, both tumor-to-tumor and cell-to-cell in single tumors. Advances in genomic technologies, especially at the single-cell level, are improving the situation, but these approaches are held back by limitations of the biotechnologies for gathering genomic data from heterogeneous cell populations and the computational methods for making sense of those data. One popular way to gain the advantages of whole-genome methods without the cost of single-cell genomics has been the use of computational deconvolution (unmixing) methods to reconstruct clonal heterogeneity from bulk genomic data. These methods, too, are limited by the difficulty of inferring genomic profiles of rare or subtly varying clonal subpopulations from bulk data, a problem that can be computationally reduced to that of reconstructing the geometry of point clouds of tumor samples in a genome space. Here, we present a new method to improve that reconstruction by better identifying subspaces corresponding to tumors produced from mixtures of distinct combinations of clonal subpopulations. We develop a nonparametric clustering method based on medoidshift clustering for identifying subgroups of tumors expected to correspond to distinct trajectories of evolutionary progression. We show on synthetic and real tumor copy-number data that this new method substantially improves our ability to resolve discrete tumor subgroups, a key step in the process of accurately deconvolving tumor genomic data and inferring clonal heterogeneity from bulk data

    Statistical Methods For Genomic And Transcriptomic Sequencing

    Get PDF
    Part 1: High-throughput sequencing of DNA coding regions has become a common way of assaying genomic variation in the study of human diseases. Copy number variation (CNV) is an important type of genomic variation, but CNV profiling from whole-exome sequencing (WES) is challenging due to the high level of biases and artifacts. We propose CODEX, a normalization and CNV calling procedure for WES data. CODEX includes a Poisson latent factor model, which includes terms that specifically remove biases due to GC content, exon capture and amplification efficiency, and latent systemic artifacts. CODEX also includes a Poisson likelihood-based segmentation procedure that explicitly models the count-based WES data. CODEX is compared to existing methods on germline CNV detection in HapMap samples using microarray-based gold standard and is further evaluated on 222 neuroblastoma samples with matched normal, with focus on somatic CNVs within the ATRX gene. Part 2: Cancer is a disease driven by evolutionary selection on somatic genetic and epigenetic alterations. We propose Canopy, a method for inferring the evolutionary phylogeny of a tumor using both somatic copy number alterations and single nucleotide alterations from one or more samples derived from a single patient. Canopy is applied to bulk sequencing datasets of both longitudinal and spatial experimental designs and to a transplantable metastasis model derived from human cancer cell line MDA-MB-231. Canopy successfully identifies cell populations and infers phylogenies that are in concordance with existing knowledge and ground truth. Through simulations, we explore the effects of key parameters on deconvolution accuracy, and compare against existing methods. Part 3: Allele-specific expression is traditionally studied by bulk RNA sequencing, which measures average expression across cells. Single-cell RNA sequencing (scRNA-seq) allows the comparison of expression distribution between the two alleles of a diploid organism and thus the characterization of allele-specific bursting. We propose SCALE to analyze genome-wide allele-specific bursting, with adjustment of technical variability. SCALE detects genes exhibiting allelic differences in bursting parameters, and genes whose alleles burst non-independently. We apply SCALE to mouse blastocyst and human fibroblast cells and find that, globally, cis control in gene expression overwhelmingly manifests as differences in burst frequency

    Characterization of cell type-specific molecular heterogeneity in cancer using multi-omic approaches

    Get PDF
    Tumors are composed of heterogeneous cell types each with its own unique molecular profiles. Recent advances in single cell genomics technologies have begun to increase our understanding of the molecular heterogeneity that exists in tumors with particular focus on gene expression and chromatin accessibility profiles. However, due to limitations in methods for certain sample types and high cost for single cell genomics, bulk tumor molecular profiling has been and remains widely used. In addition, other facets of single cell epigenomic profiling, particularly methylation and hydroxymethylation, remains underexplored. Thus, investigations to understand the cell type specific epigenetic heterogeneity and the cooperation among various molecular layers to regulate tumorigenesis are needed. In this thesis, I utilize a multi-omic approach integrating DNA methylation, hydroxymethylation, chromatin accessibility, and gene expression profiles to investigate unique single cell type-specific features in 1) epithelial-to-mesenchymal transition and in 2) pediatric central nervous system tumors. First, I demonstrate the shared and distinct epigenetic profiles that are associated with single cells undergoing epithelial-to-mesenchymal transition. With a multi-omic approach, I identify increased hydroxymethylation in binding motifs of transcription factors critical in regulating epithelial-to-mesenchymal transition. Then, I shift my focus to characterize the cellular heterogeneity in pediatric central nervous system tumors and transcriptomic alterations associated with these tumors, while accounting for cell type composition, with single nuclei gene expression data. I detect novel pediatric central nervous system tumor associated genes that are differentially expressed. Finally, I illustrate the cytosine modification alterations that occur predominantly in the progenitorlike cell types of pediatric central nervous system tumors with a multi-omic approach. I determine associations between cell type-specific hydroxymethylation alterations with cell type-specific gene expression changes. Together, these findings emphasize the need for consideration of cellular identity to determine molecular heterogeneity that exist in various cancer contexts. Moreover, these works collectively suggest the utility of multiomic approaches to uncover novel insights in underlying tumor biology

    Computational solutions for addressing heterogeneity in DNA methylation data

    Get PDF
    DNA methylation, a reversible epigenetic modification, has been implicated with various bi- ological processes including gene regulation. Due to the multitude of datasets available, it is a premier candidate for computational tool development, especially for investigating hetero- geneity within and across samples. We differentiate between three levels of heterogeneity in DNA methylation data: between-group, between-sample, and within-sample heterogeneity. Here, we separately address these three levels and present new computational approaches to quantify and systematically investigate heterogeneity. Epigenome-wide association studies relate a DNA methylation aberration to a phenotype and therefore address between-group heterogeneity. To facilitate such studies, which necessar- ily include data processing, exploratory data analysis, and differential analysis of DNA methy- lation, we extended the R-package RnBeads. We implemented novel methods for calculating the epigenetic age of individuals, novel imputation methods, and differential variability analysis. A use-case of the new features is presented using samples from Ewing sarcoma patients. As an important driver of epigenetic differences between phenotypes, we systematically investigated associations between donor genotypes and DNA methylation states in methylation quantitative trait loci (methQTL). To that end, we developed a novel computational framework –MAGAR– for determining statistically significant associations between genetic and epigenetic variations. We applied the new pipeline to samples obtained from sorted blood cells and complex bowel tissues of healthy individuals and found that tissue-specific and common methQTLs have dis- tinct genomic locations and biological properties. To investigate cell-type-specific DNA methylation profiles, which are the main drivers of within-group heterogeneity, computational deconvolution methods can be used to dissect DNA methylation patterns into latent methylation components. Deconvolution methods require pro- files of high technical quality and the identified components need to be biologically interpreted. We developed a computational pipeline to perform deconvolution of complex DNA methyla- tion data, which implements crucial data processing steps and facilitates result interpretation. We applied the protocol to lung adenocarcinoma samples and found indications of tumor in- filtration by immune cells and associations of the detected components with patient survival. Within-sample heterogeneity (WSH), i.e., heterogeneous DNA methylation patterns at a ge- nomic locus within a biological sample, is often neglected in epigenomic studies. We present the first systematic benchmark of scores quantifying WSH genome-wide using simulated and experimental data. Additionally, we created two novel scores that quantify DNA methyla- tion heterogeneity at single CpG resolution with improved robustness toward technical biases. WSH scores describe different types of WSH in simulated data, quantify differential hetero- geneity, and serve as a reliable estimator of tumor purity. Due to the broad availability of DNA methylation data, the levels of heterogeneity in DNA methylation data can be comprehensively investigated. We contribute novel computational frameworks for analyzing DNA methylation data with respect to different levels of hetero- geneity. We envision that this toolbox will be indispensible for understanding the functional implications of DNA methylation patterns in health and disease.DNA Methylierung ist eine reversible, epigenetische Modifikation, die mit verschiedenen biologischen Prozessen wie beispielsweise der Genregulation in Verbindung steht. Eine Vielzahl von DNA Methylierungsdatensätzen bildet die perfekte Grundlage zur Entwicklung von Softwareanwendungen, insbesondere um Heterogenität innerhalb und zwischen Proben zu beschreiben. Wir unterscheiden drei Ebenen von Heterogenität in DNA Methylierungsdaten: zwischen Gruppen, zwischen Proben und innerhalb einer Probe. Hier betrachten wir die drei Ebenen von Heterogenität in DNA Methylierungsdaten unabhängig voneinander und präsentieren neue Ansätze um die Heterogenität zu beschreiben und zu quantifizieren. Epigenomweite Assoziationsstudien verknüpfen eine DNA Methylierungsveränderung mit einem Phänotypen und beschreiben Heterogenität zwischen Gruppen. Um solche Studien, welche Datenprozessierung, sowie exploratorische und differentielle Datenanalyse beinhalten, zu vereinfachen haben wir die R-basierte Softwareanwendung RnBeads erweitert. Die Erweiterungen beinhalten neue Methoden, um das epigenetische Alter vorherzusagen, neue Schätzungsmethoden für fehlende Datenpunkte und eine differentielle Variabilitätsanalyse. Die Analyse von Ewing-Sarkom Patientendaten wurde als Anwendungsbeispiel für die neu entwickelten Methoden gewählt. Wir untersuchten Assoziationen zwischen Genotypen und DNA Methylierung von einzelnen CpGs, um sogenannte methylation quantitative trait loci (methQTL) zu definieren. Diese stellen einen wichtiger Faktor dar, der epigenetische Unterschiede zwischen Gruppen induziert. Hierzu entwickelten wir ein neues Softwarepaket (MAGAR), um statistisch signifikante Assoziationen zwischen genetischer und epigenetischer Variation zu identifizieren. Wir wendeten diese Pipeline auf Blutzelltypen und komplexe Biopsien von gesunden Individuen an und konnten gemeinsame und gewebespezifische methQTLs in verschiedenen Bereichen des Genoms lokalisieren, die mit unterschiedlichen biologischen Eigenschaften verknüpft sind. Die Hauptursache für Heterogenität innerhalb einer Gruppe sind zelltypspezifische DNA Methylierungsmuster. Um diese genauer zu untersuchen kann Dekonvolutionssoftware die DNA Methylierungsmatrix in unabhängige Variationskomponenten zerlegen. Dekonvolutionsmethoden auf Basis von DNA Methylierung benötigen technisch hochwertige Profile und die identifizierten Komponenten müssen biologisch interpretiert werden. In dieser Arbeit entwickelten wir eine computerbasierte Pipeline zur Durchführung von Dekonvolutionsexperimenten, welche die Datenprozessierung und Interpretation der Resultate beinhaltet. Wir wendeten das entwickelte Protokoll auf Lungenadenokarzinome an und fanden Anzeichen für eine Tumorinfiltration durch Immunzellen, sowie Verbindungen zum Überleben der Patienten. Heterogenität innerhalb einer Probe (within-sample heterogeneity, WSH), d.h. heterogene Methylierungsmuster innerhalb einer Probe an einer genomischen Position, wird in epigenomischen Studien meist vernachlässigt. Wir präsentieren den ersten Vergleich verschiedener, genomweiter WSH Maße auf simulierten und experimentellen Daten. Zusätzlich entwickelten wir zwei neue Maße um WSH für einzelne CpGs zu berechnen, welche eine verbesserte Robustheit gegenüber technischen Faktoren aufweisen. WSH Maße beschreiben verschiedene Arten von WSH, quantifizieren differentielle Heterogenität und sagen Tumorreinheit vorher. Aufgrund der breiten Verfügbarkeit von DNA Methylierungsdaten können die Ebenen der Heterogenität ganzheitlich beschrieben werden. In dieser Arbeit präsentieren wir neue Softwarelösungen zur Analyse von DNA Methylierungsdaten in Bezug auf die verschiedenen Ebenen der Heterogenität. Wir sind davon überzeugt, dass die vorgestellten Softwarewerkzeuge unverzichtbar für das Verständnis von DNA Methylierung im kranken und gesunden Stadium sein werden

    Rapid evolution and biogeographic spread in a colorectal cancer

    Get PDF
    How and when tumoral clones start spreading to surrounding and distant tissues is currently unclear. Here we leveraged a model-based evolutionary framework to investigate the demographic and biogeographic history of a colorectal cancer. Our analyses strongly support an early monoclonal metastatic colonization, followed by a rapid population expansion at both primary and secondary sites. Moreover, we infer a hematogenous metastatic spread under positive selection, plus the return of some tumoral cells from the liver back to the colon lymph nodes. This study illustrates how sophisticated techniques typical of organismal evolution can provide a detailed, quantitative picture of the complex tumoral dynamics over time and spaceEuropean Research Council | Ref. ERC-617457- PHYLOCANCERMinisterio de Economía y Competitividad | Ref. BFU2015-63774-PInstituto de Salud Carlos III | Ref. PI15/01501-FEDE
    corecore