Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 178(4): 933-948.e14, 2019 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-31398344

RESUMEN

Interferon-gamma (IFNG) augments immune function yet promotes T cell exhaustion through PDL1. How these opposing effects are integrated to impact immune checkpoint blockade (ICB) is unclear. We show that while inhibiting tumor IFNG signaling decreases interferon-stimulated genes (ISGs) in cancer cells, it increases ISGs in immune cells by enhancing IFNG produced by exhausted T cells (TEX). In tumors with favorable antigenicity, these TEX mediate rejection. In tumors with neoantigen or MHC-I loss, TEX instead utilize IFNG to drive maturation of innate immune cells, including a PD1+TRAIL+ ILC1 population. By disabling an inhibitory circuit impacting PD1 and TRAIL, blocking tumor IFNG signaling promotes innate immune killing. Thus, interferon signaling in cancer cells and immune cells oppose each other to establish a regulatory relationship that limits both adaptive and innate immune killing. In melanoma and lung cancer patients, perturbation of this relationship is associated with ICB response independent of tumor mutational burden.


Asunto(s)
Inmunidad Adaptativa/inmunología , Inmunidad Innata/inmunología , Interferón gamma/genética , Interferón gamma/metabolismo , Neoplasias Pulmonares/inmunología , Melanoma/inmunología , Traslado Adoptivo , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Linfocitos T CD8-positivos/inmunología , Antígeno CTLA-4/antagonistas & inhibidores , Línea Celular Tumoral , Estudios de Cohortes , Femenino , Técnicas de Inactivación de Genes , Humanos , Interferón gamma/antagonistas & inhibidores , Células Asesinas Naturales/inmunología , Neoplasias Pulmonares/tratamiento farmacológico , Melanoma/tratamiento farmacológico , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Supervivencia sin Progresión , RNA-Seq , Transfección
2.
Immunity ; 55(4): 671-685.e10, 2022 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-35417675

RESUMEN

Interferon-gamma (IFN-γ) has pleiotropic effects on cancer immune checkpoint blockade (ICB), including roles in ICB resistance. We analyzed gene expression in ICB-sensitive versus ICB-resistant tumor cells and identified a strong association between interferon-mediated resistance and expression of Ripk1, a regulator of tumor necrosis factor (TNF) superfamily receptors. Genetic interaction screening revealed that in cancer cells, RIPK1 diverted TNF signaling through NF-κB and away from its role in cell death. This promoted an immunosuppressive chemokine program by cancer cells, enhanced cancer cell survival, and decreased infiltration of T and NK cells expressing TNF superfamily ligands. Deletion of RIPK1 in cancer cells compromised chemokine secretion, decreased ARG1+ suppressive myeloid cells linked to ICB failure in mice and humans, and improved ICB response driven by CASP8-killing and dependent on T and NK cells. RIPK1-mediated resistance required its ubiquitin scaffolding but not kinase function. Thus, cancer cells co-opt RIPK1 to promote cell-intrinsic and cell-extrinsic resistance to immunotherapy.


Asunto(s)
Resistencia a Antineoplásicos , Inhibidores de Puntos de Control Inmunológico , Interferones , Neoplasias , Proteína Serina-Treonina Quinasas de Interacción con Receptores , Animales , Inmunoterapia , Interferón gamma/metabolismo , Interferones/metabolismo , Ratones , FN-kappa B/metabolismo , Neoplasias/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo
3.
Nature ; 619(7970): 572-584, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37468586

RESUMEN

The intestine is a complex organ that promotes digestion, extracts nutrients, participates in immune surveillance, maintains critical symbiotic relationships with microbiota and affects overall health1. The intesting has a length of over nine metres, along which there are differences in structure and function2. The localization of individual cell types, cell type development trajectories and detailed cell transcriptional programs probably drive these differences in function. Here, to better understand these differences, we evaluated the organization of single cells using multiplexed imaging and single-nucleus RNA and open chromatin assays across eight different intestinal sites from nine donors. Through systematic analyses, we find cell compositions that differ substantially across regions of the intestine and demonstrate the complexity of epithelial subtypes, and find that the same cell types are organized into distinct neighbourhoods and communities, highlighting distinct immunological niches that are present in the intestine. We also map gene regulatory differences in these cells that are suggestive of a regulatory differentiation cascade, and associate intestinal disease heritability with specific cell types. These results describe the complexity of the cell composition, regulation and organization for this organ, and serve as an important reference map for understanding human biology and disease.


Asunto(s)
Intestinos , Análisis de la Célula Individual , Humanos , Diferenciación Celular/genética , Cromatina/genética , Células Epiteliales/citología , Células Epiteliales/metabolismo , Regulación de la Expresión Génica , Mucosa Intestinal/citología , Intestinos/citología , Intestinos/inmunología , Análisis de Expresión Génica de una Sola Célula
4.
Proc Natl Acad Sci U S A ; 120(32): e2303647120, 2023 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-37523521

RESUMEN

Multimodal single-cell technologies profile multiple modalities for each cell simultaneously, enabling a more thorough characterization of cell populations. Existing dimension-reduction methods for multimodal data capture the "union of information," producing a lower-dimensional embedding that combines the information across modalities. While these tools are useful, we focus on a fundamentally different task of separating and quantifying the information among cells that is shared between the two modalities as well as unique to only one modality. Hence, we develop Tilted Canonical Correlation Analysis (Tilted-CCA), a method that decomposes a paired multimodal dataset into three lower-dimensional embeddings-one embedding captures the "intersection of information," representing the geometric relations among the cells that is common to both modalities, while the remaining two embeddings capture the "distinct information for a modality," representing the modality-specific geometric relations. We analyze single-cell multimodal datasets sequencing RNA along surface antibodies (i.e., CITE-seq) as well as RNA alongside chromatin accessibility (i.e., 10x) for blood cells and developing neurons via Tilted-CCA. These analyses show that Tilted-CCA enables meaningful visualization and quantification of the cross-modal information. Finally, Tilted-CCA's framework allows us to perform two specific downstream analyses. First, for single-cell datasets that simultaneously profile transcriptome and surface antibody markers, we show that Tilted-CCA helps design the target antibody panel to complement the transcriptome best. Second, for developmental single-cell datasets that simultaneously profile transcriptome and chromatin accessibility, we show that Tilted-CCA helps identify development-informative genes and distinguish between transient versus terminal cell types.


Asunto(s)
Algoritmos , Análisis de Correlación Canónica , Transcriptoma , Análisis de la Célula Individual/métodos
5.
PLoS Genet ; 17(6): e1009575, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34157017

RESUMEN

Over a decade of genome-wide association studies (GWAS) have led to the finding of extreme polygenicity of complex traits. The phenomenon that "all genes affect every complex trait" complicates Mendelian Randomization (MR) studies, where natural genetic variations are used as instruments to infer the causal effect of heritable risk factors. We reexamine the assumptions of existing MR methods and show how they need to be clarified to allow for pervasive horizontal pleiotropy and heterogeneous effect sizes. We propose a comprehensive framework GRAPPLE to analyze the causal effect of target risk factors with heterogeneous genetic instruments and identify possible pleiotropic patterns from data. By using GWAS summary statistics, GRAPPLE can efficiently use both strong and weak genetic instruments, detect the existence of multiple pleiotropic pathways, determine the causal direction and perform multivariable MR to adjust for confounding risk factors. With GRAPPLE, we analyze the effect of blood lipids, body mass index, and systolic blood pressure on 25 disease outcomes, gaining new information on their causal relationships and potential pleiotropic pathways involved.


Asunto(s)
Causalidad , Fenotipo , Pleiotropía Genética , Estudio de Asociación del Genoma Completo , Humanos , Análisis de la Aleatorización Mendeliana , Polimorfismo de Nucleótido Simple , Factores de Riesgo
6.
Nat Methods ; 16(9): 875-878, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31471617

RESUMEN

Single-cell RNA sequencing (scRNA-seq) data are noisy and sparse. Here, we show that transfer learning across datasets remarkably improves data quality. By coupling a deep autoencoder with a Bayesian model, SAVER-X extracts transferable gene-gene relationships across data from different labs, varying conditions and divergent species, to denoise new target datasets.


Asunto(s)
Neoplasias de la Mama/metabolismo , Biología Computacional/métodos , Leucocitos Mononucleares/metabolismo , Análisis de Secuencia de ARN/normas , Análisis de la Célula Individual/métodos , Linfocitos T/metabolismo , Transcriptoma , Animales , Teorema de Bayes , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Ratones , Análisis de Secuencia de ARN/métodos
7.
Nat Methods ; 15(7): 539-542, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29941873

RESUMEN

In single-cell RNA sequencing (scRNA-seq) studies, only a small fraction of the transcripts present in each cell are sequenced. This leads to unreliable quantification of genes with low or moderate expression, which hinders downstream analysis. To address this challenge, we developed SAVER (single-cell analysis via expression recovery), an expression recovery method for unique molecule index (UMI)-based scRNA-seq data that borrows information across genes and cells to provide accurate expression estimates for all genes.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento/métodos , ARN/genética , Análisis de la Célula Individual/métodos , Animales , Secuencia de Bases , Corteza Cerebral/citología , Perfilación de la Expresión Génica/métodos , Humanos , Ratones , ARN/química , Análisis de Secuencia de ARN/métodos , Programas Informáticos
8.
Proc Natl Acad Sci U S A ; 115(28): E6437-E6446, 2018 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-29946020

RESUMEN

Single-cell RNA sequencing (scRNA-seq) enables the quantification of each gene's expression distribution across cells, thus allowing the assessment of the dispersion, nonzero fraction, and other aspects of its distribution beyond the mean. These statistical characterizations of the gene expression distribution are critical for understanding expression variation and for selecting marker genes for population heterogeneity. However, scRNA-seq data are noisy, with each cell typically sequenced at low coverage, thus making it difficult to infer properties of the gene expression distribution from raw counts. Based on a reexamination of nine public datasets, we propose a simple technical noise model for scRNA-seq data with unique molecular identifiers (UMI). We develop deconvolution of single-cell expression distribution (DESCEND), a method that deconvolves the true cross-cell gene expression distribution from observed scRNA-seq counts, leading to improved estimates of properties of the distribution such as dispersion and nonzero fraction. DESCEND can adjust for cell-level covariates such as cell size, cell cycle, and batch effects. DESCEND's noise model and estimation accuracy are further evaluated through comparisons to RNA FISH data, through data splitting and simulations and through its effectiveness in removing known batch effects. We demonstrate how DESCEND can clarify and improve downstream analyses such as finding differentially expressed genes, identifying cell types, and selecting differentiation markers.


Asunto(s)
Regulación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Modelos Genéticos , ARN/biosíntesis , ARN/genética , Animales , Humanos
9.
Brief Bioinform ; 19(5): 731-736, 2018 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-28159966

RESUMEN

Currently, there is a lack of software for detecting copy number variations and constructing copy number profile for the whole genome from single-cell DNA sequencing data, which are often of low coverage and high technical noises. Here we introduce a new toolkit, SCNV, which features an efficient bin-free segmentation approach and provides the highest resolution possible for breakpoint detection and the subsequent copy number calling. SCNV can auto-tune parameters based on a set of normal cells from the same batch to adjust for the technical noise level of the data, facilitating its application to data gathered from different platforms and different studies.


Asunto(s)
Variaciones en el Número de Copia de ADN , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Análisis de Secuencia de ADN/métodos , Programas Informáticos , Biología Computacional/métodos , ADN de Neoplasias/genética , Genoma Humano , Secuenciación de Nucleótidos de Alto Rendimiento/estadística & datos numéricos , Humanos , Neoplasias/genética , Ploidias , Análisis de Secuencia de ADN/estadística & datos numéricos , Análisis de la Célula Individual/métodos , Análisis de la Célula Individual/estadística & datos numéricos
10.
Nucleic Acids Res ; 46(4): e19, 2018 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-29186506

RESUMEN

Large genomic rearrangements involve inversions, deletions and other structural changes that span Megabase segments of the human genome. This category of genetic aberration is the cause of many hereditary genetic disorders and contributes to pathogenesis of diseases like cancer. We developed a new algorithm called ZoomX for analysing barcode-linked sequence reads-these sequences can be traced to individual high molecular weight DNA molecules (>50 kb). To generate barcode linked sequence reads, we employ a library preparation technology (10X Genomics) that uses droplets to partition and barcode DNA molecules. Using linked read data from whole genome sequencing, we identify large genomic rearrangements, typically greater than 200kb, even when they are only present in low allelic fractions. Our algorithm uses a Poisson scan statistic to identify genomic rearrangement junctions, determine counts of junction-spanning molecules and calculate a Fisher's exact test for determining statistical significance for somatic aberrations. Utilizing a well-characterized human genome, we benchmarked this approach to accurately identify large rearrangement. Subsequently, we demonstrated that our algorithm identifies somatic rearrangements when present in lower allelic fractions as occurs in tumors. We characterized a set of complex cancer rearrangements with multiple classes of structural aberrations and with possible roles in oncogenesis.


Asunto(s)
Variación Estructural del Genoma , Neoplasias/genética , Secuenciación Completa del Genoma/métodos , Algoritmos , Aberraciones Cromosómicas , Neoplasias Gastrointestinales/genética , Genoma Humano , Humanos
11.
Bioinformatics ; 34(12): 2126-2128, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29415173

RESUMEN

Summary: Copy number variation is an important and abundant source of variation in the human genome, which has been associated with a number of diseases, especially cancer. Massively parallel next-generation sequencing allows copy number profiling with fine resolution. Such efforts, however, have met with mixed successes, with setbacks arising partly from the lack of reliable analytical methods to meet the diverse and unique challenges arising from the myriad experimental designs and study goals in genetic studies. In cancer genomics, detection of somatic copy number changes and profiling of allele-specific copy number (ASCN) are complicated by experimental biases and artifacts as well as normal cell contamination and cancer subclone admixture. Furthermore, careful statistical modeling is warranted to reconstruct tumor phylogeny by both somatic ASCN changes and single nucleotide variants. Here we describe a flexible computational pipeline, MARATHON, which integrates multiple related statistical software for copy number profiling and downstream analyses in disease genetic studies. Availability and implementation: MARATHON is publicly available at https://github.com/yuchaojiang/MARATHON. Supplementary information: Supplementary data are available at Bioinformatics online.


Asunto(s)
Variaciones en el Número de Copia de ADN , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Modelos Estadísticos , Neoplasias/genética , Análisis de Secuencia de ADN/métodos , Programas Informáticos , Algoritmos , Genómica/métodos , Humanos , Modelos Genéticos , Polimorfismo de Nucleótido Simple
12.
Nucleic Acids Res ; 45(19): 10978-10988, 2017 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-29036714

RESUMEN

Recent technological breakthroughs have made it possible to measure RNA expression at the single-cell level, thus paving the way for exploring expression heterogeneity among individual cells. Current single-cell RNA sequencing (scRNA-seq) protocols are complex and introduce technical biases that vary across cells, which can bias downstream analysis without proper adjustment. To account for cell-to-cell technical differences, we propose a statistical framework, TASC (Toolkit for Analysis of Single Cell RNA-seq), an empirical Bayes approach to reliably model the cell-specific dropout rates and amplification bias by use of external RNA spike-ins. TASC incorporates the technical parameters, which reflect cell-to-cell batch effects, into a hierarchical mixture model to estimate the biological variance of a gene and detect differentially expressed genes. More importantly, TASC is able to adjust for covariates to further eliminate confounding that may originate from cell size and cell cycle differences. In simulation and real scRNA-seq data, TASC achieves accurate Type I error control and displays competitive sensitivity and improved robustness to batch effects in differential expression analysis, compared to existing methods. TASC is programmed to be computationally efficient, taking advantage of multi-threaded parallelization. We believe that TASC will provide a robust platform for researchers to leverage the power of scRNA-seq.


Asunto(s)
Biología Computacional/métodos , Perfilación de la Expresión Génica/métodos , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Algoritmos , Teorema de Bayes , Reproducibilidad de los Resultados , Programas Informáticos
13.
Proc Natl Acad Sci U S A ; 113(37): E5528-37, 2016 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-27573852

RESUMEN

Cancer is a disease driven by evolutionary selection on somatic genetic and epigenetic alterations. Here, 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. Canopy is an open-source R package available at https://cran.r-project.org/web/packages/Canopy/.


Asunto(s)
Evolución Clonal/genética , Evolución Molecular , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Neoplasias/genética , Biología Computacional/métodos , Variaciones en el Número de Copia de ADN/genética , Exoma/genética , Heterogeneidad Genética , Humanos , Internet , Mutación , Neoplasias/patología , Filogenia , Polimorfismo de Nucleótido Simple , Programas Informáticos
14.
Biometrics ; 74(4): 1150-1160, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29603714

RESUMEN

In observational studies to estimate treatment effects, unmeasured confounding is often a concern. The instrumental variable (IV) method can control for unmeasured confounding when there is a valid IV. To be a valid IV, a variable needs to be independent of unmeasured confounders and only affect the outcome through affecting the treatment. When applying the IV method, there is often concern that a putative IV is invalid to some degree. We present an approach to sensitivity analysis for the IV method which examines the sensitivity of inferences to violations of IV validity. Specifically, we consider sensitivity when the magnitude of association between the putative IV and the unmeasured confounders and the direct effect of the IV on the outcome are limited in magnitude by a sensitivity parameter. Our approach is based on extending the Anderson-Rubin test and is valid regardless of the strength of the instrument. A power formula for this sensitivity analysis is presented. We illustrate its usage via examples about Mendelian randomization studies and its implications via a comparison of using rare versus common genetic variants as instruments.


Asunto(s)
Biometría/métodos , Factores de Confusión Epidemiológicos , Análisis de la Aleatorización Mendeliana/métodos , Análisis de Causa Raíz/métodos , Sesgo , Variación Genética , Humanos , Estudios Observacionales como Asunto , Evaluación de Resultado en la Atención de Salud
15.
Nucleic Acids Res ; 44(15): e126, 2016 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-27325742

RESUMEN

We present SWAN, a statistical framework for robust detection of genomic structural variants in next-generation sequencing data and an analysis of mid-range size insertion and deletions (<10 Kb) for whole genome analysis and DNA mixtures. To identify these mid-range size events, SWAN collectively uses information from read-pair, read-depth and one end mapped reads through statistical likelihoods based on Poisson field models. SWAN also uses soft-clip/split read remapping to supplement the likelihood analysis and determine variant boundaries. The accuracy of SWAN is demonstrated by in silico spike-ins and by identification of known variants in the NA12878 genome. We used SWAN to identify a series of novel set of mid-range insertion/deletion detection that were confirmed by targeted deep re-sequencing. An R package implementation of SWAN is open source and freely available.


Asunto(s)
Análisis Mutacional de ADN/métodos , Genoma/genética , Genómica/métodos , Mutación INDEL/genética , Adenoviridae/genética , Algoritmos , Animales , Benchmarking , Simulación por Computador , Conjuntos de Datos como Asunto , Pan troglodytes/virología , Distribución de Poisson , Reproducibilidad de los Resultados
16.
Bioinformatics ; 32(6): 926-8, 2016 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-26576652

RESUMEN

UNLABELLED: In this article, we introduce a robust and efficient strategy for deriving global and allele-specific copy number alternations (CNA) from cancer whole exome sequencing data based on Log R ratios and B-allele frequencies. Applying the approach to the analysis of over 200 skin cancer samples, we demonstrate its utility for discovering distinct CNA events and for deriving ancillary information such as tumor purity. AVAILABILITY AND IMPLEMENTATION: https://github.com/xfwang/CLOSE CONTACT: xuefeng.wang@stonybrook.edu or michael.krauthammer@yale.edu SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Variaciones en el Número de Copia de ADN , Algoritmos , Frecuencia de los Genes , Genoma Humano , Humanos , Neoplasias , Análisis de Secuencia de ADN
17.
Nucleic Acids Res ; 43(6): e39, 2015 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-25618849

RESUMEN

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 detecting and characterizing CNV from exome sequencing is challenging due to the high level of biases and artifacts. We propose CODEX, a normalization and CNV calling procedure for whole exome sequencing data. The Poisson latent factor model in CODEX 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 recursive segmentation procedure that explicitly models the count-based exome sequencing data. CODEX is compared to existing methods on a population analysis of HapMap samples from the 1000 Genomes Project, and shown to be more accurate on three microarray-based validation data sets. We further evaluate performance on 222 neuroblastoma samples with matched normals and focus on a well-studied rare somatic CNV within the ATRX gene. We show that the cross-sample normalization procedure of CODEX removes more noise than normalizing the tumor against the matched normal and that the segmentation procedure performs well in detecting CNVs with nested structures.


Asunto(s)
Variaciones en el Número de Copia de ADN , Exoma , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Análisis de Secuencia de ADN/métodos , Algoritmos , Composición de Base , Sesgo , Estudios de Casos y Controles , ADN Helicasas/genética , ADN de Neoplasias/genética , Bases de Datos de Ácidos Nucleicos/estadística & datos numéricos , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento/estadística & datos numéricos , Humanos , Funciones de Verosimilitud , Masculino , Neuroblastoma/genética , Proteínas Nucleares/genética , Análisis de Secuencia de ADN/estadística & datos numéricos , Proteína Nuclear Ligada al Cromosoma X
18.
Nucleic Acids Res ; 43(4): e23, 2015 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-25477383

RESUMEN

The progression and clonal development of tumors often involve amplifications and deletions of genomic DNA. Estimation of allele-specific copy number, which quantifies the number of copies of each allele at each variant loci rather than the total number of chromosome copies, is an important step in the characterization of tumor genomes and the inference of their clonal history. We describe a new method, falcon, for finding somatic allele-specific copy number changes by next generation sequencing of tumors with matched normals. falcon is based on a change-point model on a bivariate mixed Binomial process, which explicitly models the copy numbers of the two chromosome haplotypes and corrects for local allele-specific coverage biases. By using the Binomial distribution rather than a normal approximation, falcon more effectively pools evidence from sites with low coverage. A modified Bayesian information criterion is used to guide model selection for determining the number of copy number events. Falcon is evaluated on in silico spike-in data and applied to the analysis of a pre-malignant colon tumor sample and late-stage colorectal adenocarcinoma from the same individual. The allele-specific copy number estimates obtained by falcon allows us to draw detailed conclusions regarding the clonal history of the individual's colon cancer.


Asunto(s)
Alelos , Dosificación de Gen , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Neoplasias/genética , Análisis de Secuencia de ADN/métodos , Programas Informáticos , Adenocarcinoma/genética , Evolución Clonal , Neoplasias Colorrectales/genética , Humanos
19.
BMC Genomics ; 16 Suppl 5: S5, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26040834

RESUMEN

BACKGROUND: A fundamental question in neuroscience is how memories are stored and retrieved in the brain. Long-term memory formation requires transcription, translation and epigenetic processes that control gene expression. Thus, characterizing genome-wide the transcriptional changes that occur after memory acquisition and retrieval is of broad interest and importance. Genome-wide technologies are commonly used to interrogate transcriptional changes in discovery-based approaches. Their ability to increase scientific insight beyond traditional candidate gene approaches, however, is usually hindered by batch effects and other sources of unwanted variation, which are particularly hard to control in the study of brain and behavior. RESULTS: We examined genome-wide gene expression after contextual conditioning in the mouse hippocampus, a brain region essential for learning and memory, at all the time-points in which inhibiting transcription has been shown to impair memory formation. We show that most of the variance in gene expression is not due to conditioning and that by removing unwanted variance through additional normalization we are able provide novel biological insights. In particular, we show that genes downregulated by memory acquisition and retrieval impact different functions: chromatin assembly and RNA processing, respectively. Levels of histone 2A variant H2AB are reduced only following acquisition, a finding we confirmed using quantitative proteomics. On the other hand, splicing factor Rbfox1 and NMDA receptor-dependent microRNA miR-219 are only downregulated after retrieval, accompanied by an increase in protein levels of miR-219 target CAMKIIγ. CONCLUSIONS: We provide a thorough characterization of coding and non-coding gene expression during long-term memory formation. We demonstrate that unwanted variance dominates the signal in transcriptional studies of learning and memory and introduce the removal of unwanted variance through normalization as a necessary step for the analysis of genome-wide transcriptional studies in the context of brain and behavior. We show for the first time that histone variants are downregulated after memory acquisition, and splicing factors and microRNAs after memory retrieval. Our results provide mechanistic insights into the molecular basis of cognition by highlighting the differential involvement of epigenetic mechanisms, such as histone variants and post-transcriptional RNA regulation, after acquisition and retrieval of memory.


Asunto(s)
Epigénesis Genética/fisiología , Hipocampo/fisiología , Histonas/genética , Memoria a Largo Plazo/fisiología , MicroARNs/genética , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Condicionamiento Psicológico/fisiología , Expresión Génica , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Masculino , Memoria a Corto Plazo/fisiología , Ratones , Ratones Endogámicos C57BL , MicroARNs/biosíntesis , Factores de Empalme de ARN , Proteínas de Unión al ARN/genética , Transcripción Genética/genética
20.
Circ Res ; 122(12): 1632-1634, 2018 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-29880495
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA