5,507 research outputs found

    Phenotypic heterogeneity in modeling cancer evolution

    Full text link
    The unwelcome evolution of malignancy during cancer progression emerges through a selection process in a complex heterogeneous population structure. In the present work, we investigate evolutionary dynamics in a phenotypically heterogeneous population of stem cells (SCs) and their associated progenitors. The fate of a malignant mutation is determined not only by overall stem cell and differentiated cell growth rates but also differentiation and dedifferentiation rates. We investigate the effect of such a complex population structure on the evolution of malignant mutations. We derive exact analytic results for the fixation probability of a mutant arising in each of the subpopulations. The analytic results are in almost perfect agreement with the numerical simulations. Moreover, a condition for evolutionary advantage of a mutant cell versus the wild type population is given in the present study. We also show that microenvironment-induced plasticity in invading mutants leads to more aggressive mutants with higher fixation probability. Our model predicts that decreasing polarity between stem and differentiated cells turnover would raise the survivability of non-plastic mutants; while it would suppress the development of malignancy for plastic mutants. We discuss our model in the context of colorectal/intestinal cancer (at the epithelium). This novel mathematical framework can be applied more generally to a variety of problems concerning selection in heterogeneous populations, in other contexts such as population genetics, and ecology.Comment: 28 pages, 7 figures, 2 table

    A systematic approach to cancer: evolution beyond selection.

    Get PDF
    Cancer is typically scrutinized as a pathological process characterized by chromosomal aberrations and clonal expansion subject to stochastic Darwinian selection within adaptive cellular ecosystems. Cognition based evolution is suggested as an alternative approach to cancer development and progression in which neoplastic cells of differing karyotypes and cellular lineages are assessed as self-referential agencies with purposive participation within tissue microenvironments. As distinct self-aware entities, neoplastic cells occupy unique participant/observer status within tissue ecologies. In consequence, neoplastic proliferation by clonal lineages is enhanced by the advantaged utilization of ecological resources through flexible re-connection with progenitor evolutionary stages

    cyTRON and cyTRON/JS: two Cytoscape-based applications for the inference of cancer evolution models

    Full text link
    The increasing availability of sequencing data of cancer samples is fueling the development of algorithmic strategies to investigate tumor heterogeneity and infer reliable models of cancer evolution. We here build up on previous works on cancer progression inference from genomic alteration data, to deliver two distinct Cytoscape-based applications, which allow to produce, visualize and manipulate cancer evolution models, also by interacting with public genomic and proteomics databases. In particular, we here introduce cyTRON, a stand-alone Cytoscape app, and cyTRON/JS, a web application which employs the functionalities of Cytoscape/JS. cyTRON was developed in Java; the code is available at https://github.com/BIMIB-DISCo/cyTRON and on the Cytoscape App Store http://apps.cytoscape.org/apps/cytron. cyTRON/JS was developed in JavaScript and R; the source code of the tool is available at https://github.com/BIMIB-DISCo/cyTRON-js and the tool is accessible from https://bimib.disco.unimib.it/cytronjs/welcome

    Single-cell epigenomic variability reveals functional cancer heterogeneity.

    Get PDF
    BackgroundCell-to-cell heterogeneity is a major driver of cancer evolution, progression, and emergence of drug resistance. Epigenomic variation at the single-cell level can rapidly create cancer heterogeneity but is difficult to detect and assess functionally.ResultsWe develop a strategy to bridge the gap between measurement and function in single-cell epigenomics. Using single-cell chromatin accessibility and RNA-seq data in K562 leukemic cells, we identify the cell surface marker CD24 as co-varying with chromatin accessibility changes linked to GATA transcription factors in single cells. Fluorescence-activated cell sorting of CD24 high versus low cells prospectively isolated GATA1 and GATA2 high versus low cells. GATA high versus low cells express differential gene regulatory networks, differential sensitivity to the drug imatinib mesylate, and differential self-renewal capacity. Lineage tracing experiments show that GATA/CD24hi cells have the capability to rapidly reconstitute the heterogeneity within the entire starting population, suggesting that GATA expression levels drive a phenotypically relevant source of epigenomic plasticity.ConclusionSingle-cell chromatin accessibility can guide prospective characterization of cancer heterogeneity. Epigenomic subpopulations in cancer impact drug sensitivity and the clonal dynamics of cancer evolution

    Traveling wave solution of the Hele-Shaw model of tumor growth with nutrient

    Get PDF
    Several mathematical models of tumor growth are now commonly used to explain medical observations and predict cancer evolution based on images. These models incorporate mechanical laws for tissue compression combined with rules for nutrients availability which can differ depending on the situation under consideration, in vivo or in vitro. Numerical solutions exhibit, as expected from medical observations, a proliferative rim and a necrotic core. However, their precise profiles are rather complex, both in one and two dimensions.Comment: 25 page

    Fast and scalable inference of multi-sample cancer lineages.

    Get PDF
    Somatic variants can be used as lineage markers for the phylogenetic reconstruction of cancer evolution. Since somatic phylogenetics is complicated by sample heterogeneity, novel specialized tree-building methods are required for cancer phylogeny reconstruction. We present LICHeE (Lineage Inference for Cancer Heterogeneity and Evolution), a novel method that automates the phylogenetic inference of cancer progression from multiple somatic samples. LICHeE uses variant allele frequencies of somatic single nucleotide variants obtained by deep sequencing to reconstruct multi-sample cell lineage trees and infer the subclonal composition of the samples. LICHeE is open source and available at http://viq854.github.io/lichee

    Epigenetic noise fuels cancer evolution

    Get PDF
    Cancer is a disease of the genome and the epigenome. Previous studies have shown that genomic changes such as mutations, copy number variation, and genomic rearrangements drive cancer evolution. In this issue of Cancer Cell, Landau and colleagues add epigenomic changes, specifically locally disordered DNA methylation, to cancer’s evolutionary trajectory

    Cancer evolution and individual susceptibility

    Get PDF
    El pdf del artículo es la versión de autor.Cancer susceptibility is due to interactions between inherited genetic factors and exposure to environmental carcinogens. The genetic component is constituted mainly by weakly acting low-penetrance genetic variants that interact among themselves, as well as with the environment. These low susceptibility genes can be categorized into two main groups: one includes those that control intrinsic tumor cell activities (i.e. apoptosis, proliferation or DNA repair), and the other contains those that modulate the function of extrinsic tumor cell compartments (i.e. stroma, angiogenesis, or endocrine and immune systems). Genome-Wide Association Studies (GWAS) of human populations have identified numerous genetic loci linked with cancer risk and behavior, but nevertheless the major component of cancer heritability remains to be explained. One reason may be that GWAS cannot readily capture gene–gene or gene–environment interactions. Mouse model approaches offer an alternative or complementary strategy, because of our ability to control both the genetic and environmental components of risk. Recently developed genetic tools, including high-throughput technologies such as SNP, CGH and gene expression microarrays, have led to more powerful strategies for refining quantitative trait loci (QTL) and identifying the critical genes. In particular, the cross-species approaches will help to refine locations of QTLs, and reveal their genetic and environmental interactions. The identification of human tumor susceptibility genes and discovery of their roles in carcinogenesis will ultimately be important for the development of methods for prediction of risk, diagnosis, prevention and therapy for human cancers.J. H. Mao is supported by Office of Biological & Environmental Research, of the U.S. Department of Energy under Contract No. DE-AC02-05CH11231, by Laboratory Downloaded on 20 February 2012 Published on 24 January 2011 on http://pubs.rsc.org | doi:10.1039/C0IB00094A View Online This journal is c The Royal Society of Chemistry 2011 Integr. Biol., 2011, 3, 316–328 325 Directed Research & Development Program (LDRD), and by the National Institutes of Health, National Cancer Institute grant R01 CA116481. J. Pe´rez-Losada is partially supported by FEDER and MICINN (PLE2009-119), FIS (PI070057; PI10/00328), CSIC (200920I137), Junta de Castilla y Leo´ n (SAN126/SA66/09; SA079A09). A. Castellanos-Martı´n is supported by FEDER and MICINN (PLE2009-119).Peer reviewe

    Mathematical modelling of cancer evolution

    Get PDF
    The term "cancer" refers to a group of diseases which can affect almost any tissue and organ and are characterised by the uncontrolled growth of abnormal cells, whose cell cycle is much faster than the one of a healthy cell. The carcinogenesis, i.e. the production of a cancer, is due to mutations that occur physiologically during the DNA replication process. From a mathematical point of view, many aspects can be studied. In this thesis, we focus on tumour initial developement, modelling the oncogenesis through a system of ordinary differential equations (ODEs), taking into account the possibility that a mutation occurs and the competition among healthy cells and cancer cells for the available resource, such as physical space to develop, nutrients and oxygen. The model is later analysed: it is done analitically when we are differentiating between healthy cells and cancer ones; otherwise, if we want to consider n different genotypes, the study is numerical. Some biological interpretation is given to the results.ope

    Cancer evolution: Darwin and beyond

    Get PDF
    Clinical and laboratory studies over recent decades have established branched evolution as a feature of cancer. However, while grounded in somatic selection, several lines of evidence suggest a Darwinian model alone is insufficient to fully explain cancer evolution. First, the role of macroevolutionary events in tumour initiation and progression contradicts Darwin's central thesis of gradualism. Whole-genome doubling, chromosomal chromoplexy and chromothripsis represent examples of single catastrophic events which can drive tumour evolution. Second, neutral evolution can play a role in some tumours, indicating that selection is not always driving evolution. Third, increasing appreciation of the role of the ageing soma has led to recent generalised theories of age-dependent carcinogenesis. Here, we review these concepts and others, which collectively argue for a model of cancer evolution which extends beyond Darwin. We also highlight clinical opportunities which can be grasped through targeting cancer vulnerabilities arising from non-Darwinian patterns of evolution
    • …
    corecore