Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
BMC Biol ; 19(1): 203, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34526021

RESUMEN

BACKGROUND: Silencing of transposable elements (TEs) is essential for maintaining genome stability. Plants use small RNAs (sRNAs) to direct DNA methylation to TEs (RNA-directed DNA methylation; RdDM). Similar mechanisms of epigenetic silencing in the fungal kingdom have remained elusive. RESULTS: We use sRNA sequencing and methylation data to gain insight into epigenetics in the dikaryotic fungus Puccinia graminis f. sp. tritici (Pgt), which causes the devastating stem rust disease on wheat. We use Hi-C data to define the Pgt centromeres and show that they are repeat-rich regions (~250 kb) that are highly diverse in sequence between haplotypes and, like in plants, are enriched for young TEs. DNA cytosine methylation is particularly active at centromeres but also associated with genome-wide control of young TE insertions. Strikingly, over 90% of Pgt sRNAs and several RNAi genes are differentially expressed during infection. Pgt induces waves of functionally diversified sRNAs during infection. The early wave sRNAs are predominantly 21 nts with a 5' uracil derived from genes. In contrast, the late wave sRNAs are mainly 22-nt sRNAs with a 5' adenine and are strongly induced from centromeric regions. TEs that overlap with late wave sRNAs are more likely to be methylated, both inside and outside the centromeres, and methylated TEs exhibit a silencing effect on nearby genes. CONCLUSIONS: We conclude that rust fungi use an epigenetic silencing pathway that might have similarity with RdDM in plants. The Pgt RNAi machinery and sRNAs are under tight temporal control throughout infection and might ensure genome stability during sporulation.


Asunto(s)
Basidiomycota , Metilación de ADN , Puccinia , Basidiomycota/genética , Centrómero , Metilación de ADN/genética , Elementos Transponibles de ADN , Inestabilidad Genómica , Humanos , Enfermedades de las Plantas/genética , Puccinia/patogenicidad , ARN
2.
Genome Res ; 25(5): 762-74, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25840857

RESUMEN

Saccharomyces cerevisiae, a well-established model for species as diverse as humans and pathogenic fungi, is more recently a model for population and quantitative genetics. S. cerevisiae is found in multiple environments-one of which is the human body-as an opportunistic pathogen. To aid in the understanding of the S. cerevisiae population and quantitative genetics, as well as its emergence as an opportunistic pathogen, we sequenced, de novo assembled, and extensively manually edited and annotated the genomes of 93 S. cerevisiae strains from multiple geographic and environmental origins, including many clinical origin strains. These 93 S. cerevisiae strains, the genomes of which are near-reference quality, together with seven previously sequenced strains, constitute a novel genetic resource, the "100-genomes" strains. Our sequencing coverage, high-quality assemblies, and annotation provide unprecedented opportunities for detailed interrogation of complex genomic loci, examples of which we demonstrate. We found most phenotypic variation to be quantitative and identified population, genotype, and phenotype associations. Importantly, we identified clinical origin associations. For example, we found that an introgressed PDR5 was present exclusively in clinical origin mosaic group strains; that the mosaic group was significantly enriched for clinical origin strains; and that clinical origin strains were much more copper resistant, suggesting that copper resistance contributes to fitness in the human host. The 100-genomes strains are a novel, multipurpose resource to advance the study of S. cerevisiae population genetics, quantitative genetics, and the emergence of an opportunistic pathogen.


Asunto(s)
Mapeo Contig/métodos , Genoma Fúngico , Genotipo , Fenotipo , Polimorfismo Genético , Saccharomyces cerevisiae/genética , Alineación de Secuencia/métodos , Filogenia , Saccharomyces cerevisiae/clasificación , Saccharomyces cerevisiae/patogenicidad , Virulencia/genética
3.
Nature ; 482(7384): 173-8, 2012 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-22318601

RESUMEN

A major challenge of biology is understanding the relationship between molecular genetic variation and variation in quantitative traits, including fitness. This relationship determines our ability to predict phenotypes from genotypes and to understand how evolutionary forces shape variation within and between species. Previous efforts to dissect the genotype-phenotype map were based on incomplete genotypic information. Here, we describe the Drosophila melanogaster Genetic Reference Panel (DGRP), a community resource for analysis of population genomics and quantitative traits. The DGRP consists of fully sequenced inbred lines derived from a natural population. Population genomic analyses reveal reduced polymorphism in centromeric autosomal regions and the X chromosome, evidence for positive and negative selection, and rapid evolution of the X chromosome. Many variants in novel genes, most at low frequency, are associated with quantitative traits and explain a large fraction of the phenotypic variance. The DGRP facilitates genotype-phenotype mapping using the power of Drosophila genetics.


Asunto(s)
Drosophila melanogaster/genética , Estudio de Asociación del Genoma Completo , Genómica , Sitios de Carácter Cuantitativo/genética , Alelos , Animales , Centrómero/genética , Cromosomas de Insectos/genética , Genotipo , Fenotipo , Polimorfismo de Nucleótido Simple/genética , Selección Genética/genética , Inanición/genética , Telómero/genética , Cromosoma X/genética
4.
Proc Natl Acad Sci U S A ; 112(44): E6010-9, 2015 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-26483487

RESUMEN

Understanding how DNA sequence variation is translated into variation for complex phenotypes has remained elusive but is essential for predicting adaptive evolution, for selecting agriculturally important animals and crops, and for personalized medicine. Gene expression may provide a link between variation in DNA sequence and organismal phenotypes, and its abundance can be measured efficiently and accurately. Here we quantified genome-wide variation in gene expression in the sequenced inbred lines of the Drosophila melanogaster Genetic Reference Panel (DGRP), increasing the annotated Drosophila transcriptome by 11%, including thousands of novel transcribed regions (NTRs). We found that 42% of the Drosophila transcriptome is genetically variable in males and females, including the NTRs, and is organized into modules of genetically correlated transcripts. We found that NTRs often were negatively correlated with the expression of protein-coding genes, which we exploited to annotate NTRs functionally. We identified regulatory variants for the mean and variance of gene expression, which have largely independent genetic control. Expression quantitative trait loci (eQTLs) for the mean, but not for the variance, of gene expression were concentrated near genes. Notably, the variance eQTLs often interacted epistatically with local variants in these genes to regulate gene expression. This comprehensive characterization of population-scale diversity of transcriptomes and its genetic basis in the DGRP is critically important for a systems understanding of quantitative trait variation.


Asunto(s)
Drosophila melanogaster/genética , Transcriptoma , Animales , Epistasis Genética , Sitios de Carácter Cuantitativo
5.
Genome Res ; 24(7): 1193-208, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24714809

RESUMEN

The Drosophila melanogaster Genetic Reference Panel (DGRP) is a community resource of 205 sequenced inbred lines, derived to improve our understanding of the effects of naturally occurring genetic variation on molecular and organismal phenotypes. We used an integrated genotyping strategy to identify 4,853,802 single nucleotide polymorphisms (SNPs) and 1,296,080 non-SNP variants. Our molecular population genomic analyses show higher deletion than insertion mutation rates and stronger purifying selection on deletions. Weaker selection on insertions than deletions is consistent with our observed distribution of genome size determined by flow cytometry, which is skewed toward larger genomes. Insertion/deletion and single nucleotide polymorphisms are positively correlated with each other and with local recombination, suggesting that their nonrandom distributions are due to hitchhiking and background selection. Our cytogenetic analysis identified 16 polymorphic inversions in the DGRP. Common inverted and standard karyotypes are genetically divergent and account for most of the variation in relatedness among the DGRP lines. Intriguingly, variation in genome size and many quantitative traits are significantly associated with inversions. Approximately 50% of the DGRP lines are infected with Wolbachia, and four lines have germline insertions of Wolbachia sequences, but effects of Wolbachia infection on quantitative traits are rarely significant. The DGRP complements ongoing efforts to functionally annotate the Drosophila genome. Indeed, 15% of all D. melanogaster genes segregate for potentially damaged proteins in the DGRP, and genome-wide analyses of quantitative traits identify novel candidate genes. The DGRP lines, sequence data, genotypes, quality scores, phenotypes, and analysis and visualization tools are publicly available.


Asunto(s)
Drosophila melanogaster/genética , Variación Genética , Genoma de los Insectos , Fenotipo , Animales , Cromatina/genética , Cromatina/metabolismo , Drosophila melanogaster/microbiología , Femenino , Ligamiento Genético , Tamaño del Genoma , Estudio de Asociación del Genoma Completo , Genotipo , Técnicas de Genotipaje , Secuenciación de Nucleótidos de Alto Rendimiento , Mutación INDEL , Desequilibrio de Ligamiento , Masculino , Anotación de Secuencia Molecular , Polimorfismo de Nucleótido Simple , Carácter Cuantitativo Heredable , Reproducibilidad de los Resultados
6.
Mol Biol Evol ; 31(2): 425-33, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24214536

RESUMEN

Gene conversion is the nonreciprocal exchange of genetic material between homologous chromosomes. Multiple lines of evidence from a variety of taxa strongly suggest that gene conversion events are biased toward GC-bearing alleles. However, in Drosophila, the data have largely been indirect and unclear, with some studies supporting the predictions of a GC-biased gene conversion model and other data showing contradictory findings. Here, we test whether gene conversion events are GC-biased in Drosophila melanogaster using whole-genome polymorphism and divergence data. Our results provide no support for GC-biased gene conversion and thus suggest that this process is unlikely to significantly contribute to patterns of polymorphism and divergence in this system.


Asunto(s)
Citosina/metabolismo , Drosophila melanogaster/genética , Conversión Génica , Guanina/metabolismo , Alelos , Animales , Cromosomas de Insectos , Evolución Molecular , Genoma de los Insectos , Genómica , Tasa de Mutación , Filogenia , Polimorfismo Genético
7.
Genome Res ; 22(5): 966-74, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22367192

RESUMEN

High-throughput sequencing is enabling remarkably deep surveys of genomic variation. It is now possible to completely sequence multiple individuals from a single species, yet the identification of variation among them remains an evolving computational challenge. This challenge is compounded for experimental organisms when strains are studied instead of individuals. In response, we present the Joint Genotyper for Inbred Lines (JGIL) as a method for obtaining genotypes and identifying variation among a large panel of inbred strains or lines. JGIL inputs the sequence reads from each line after their alignment to a common reference. Its probabilistic model includes site-specific parameters common to all lines that describe the frequency of nucleotides segregating in the population from which the inbred panel was derived. The distribution of line genotypes is conditional on these parameters and reflects the experimental design. Site-specific error probabilities, also common to all lines, parameterize the distribution of reads conditional on line genotype and realized coverage. Both sets of parameters are estimated per site from the aggregate read data, and posterior probabilities are calculated to decode the genotype of each line. We present an application of JGIL to 162 inbred Drosophila melanogaster lines from the Drosophila Genetic Reference Panel. We explore by simulation the effect of varying coverage, sequencing error, mapping error, and the number of lines. In doing so, we illustrate how JGIL is robust to moderate levels of error. Supported by these analyses, we advocate the importance of modeling the data and the experimental design when possible.


Asunto(s)
Drosophila melanogaster/genética , Variación Genética , Técnicas de Genotipaje , Algoritmos , Animales , Mapeo Cromosómico , Simulación por Computador , Técnicas de Genotipaje/normas , Endogamia , Funciones de Verosimilitud , Modelos Genéticos , Polimorfismo de Nucleótido Simple , Estándares de Referencia , Análisis de Secuencia de ADN
8.
PLoS Pathog ; 9(8): e1003574, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24009506

RESUMEN

Aflatoxins are produced by Aspergillus flavus and A. parasiticus in oil-rich seed and grain crops and are a serious problem in agriculture, with aflatoxin B1 being the most carcinogenic natural compound known. Sexual reproduction in these species occurs between individuals belonging to different vegetative compatibility groups (VCGs). We examined natural genetic variation in 758 isolates of A. flavus, A. parasiticus and A. minisclerotigenes sampled from single peanut fields in the United States (Georgia), Africa (Benin), Argentina (Córdoba), Australia (Queensland) and India (Karnataka). Analysis of DNA sequence variation across multiple intergenic regions in the aflatoxin gene clusters of A. flavus, A. parasiticus and A. minisclerotigenes revealed significant linkage disequilibrium (LD) organized into distinct blocks that are conserved across different localities, suggesting that genetic recombination is nonrandom and a global occurrence. To assess the contributions of asexual and sexual reproduction to fixation and maintenance of toxin chemotype diversity in populations from each locality/species, we tested the null hypothesis of an equal number of MAT1-1 and MAT1-2 mating-type individuals, which is indicative of a sexually recombining population. All samples were clone-corrected using multi-locus sequence typing which associates closely with VCG. For both A. flavus and A. parasiticus, when the proportions of MAT1-1 and MAT1-2 were significantly different, there was more extensive LD in the aflatoxin cluster and populations were fixed for specific toxin chemotype classes, either the non-aflatoxigenic class in A. flavus or the B1-dominant and G1-dominant classes in A. parasiticus. A mating type ratio close to 1∶1 in A. flavus, A. parasiticus and A. minisclerotigenes was associated with higher recombination rates in the aflatoxin cluster and less pronounced chemotype differences in populations. This work shows that the reproductive nature of the population (more sexual versus more asexual) is predictive of aflatoxin chemotype diversity in these agriculturally important fungi.


Asunto(s)
Aflatoxinas/biosíntesis , Aspergillus flavus/metabolismo , Proteínas Fúngicas/metabolismo , Genes Fúngicos/fisiología , Familia de Multigenes/fisiología , Proteínas Represoras/metabolismo , Aflatoxinas/genética , Aspergillus flavus/genética , Proteínas Fúngicas/genética , Proteínas Represoras/genética , Especificidad de la Especie
9.
Mol Ecol ; 24(8): 1889-909, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25773520

RESUMEN

Aspergillus flavus and A. parasiticus are the two most important aflatoxin-producing fungi responsible for the contamination of agricultural commodities worldwide. Both species are heterothallic and undergo sexual reproduction in laboratory crosses. Here we examine the possibility of interspecific matings between A. flavus and A. parasiticus. These species can be distinguished morphologically and genetically, as well as by their mycotoxin profiles. Aspergillus flavus produces both B aflatoxins and cyclopiazonic acid (CPA), B aflatoxins or CPA alone, or neither mycotoxin; Aspergillus parasiticus produces B and G aflatoxins or the aflatoxin precursor O-methylsterigmatocystin, but not CPA. Only four of forty-five attempted interspecific crosses between opposite mating types of A. flavus and A. parasiticus were fertile and produced viable ascospores. Single ascospore strains from each cross were shown to be recombinant hybrids using multilocus genotyping and array comparative genome hybridization. Conidia of parents and their hybrid progeny were haploid and predominantly monokaryons and dikaryons based on flow cytometry. Multilocus phylogenetic inference showed that experimental hybrid progeny were grouped with naturally occurring A. flavus L strain and A. parasiticus. Higher total aflatoxin concentrations in some F1 progeny strains compared to midpoint parent aflatoxin levels indicate synergism in aflatoxin production; moreover, three progeny strains synthesized G aflatoxins that were not produced by the parents, and there was evidence of allopolyploidization in one strain. These results suggest that hybridization is an important diversifying force resulting in the genesis of novel toxin profiles in these agriculturally important fungi.


Asunto(s)
Aflatoxinas/biosíntesis , Aspergillus flavus/genética , Aspergillus/genética , Hibridación Genética , Aspergillus/clasificación , Hibridación Genómica Comparativa , Genes del Tipo Sexual de los Hongos , Genotipo , Técnicas de Genotipaje , Datos de Secuencia Molecular , Fenotipo , Filogenia , Análisis de Secuencia de ADN , Esterigmatocistina/análogos & derivados , Esterigmatocistina/biosíntesis
10.
Nat Rev Genet ; 10(8): 565-77, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19584810

RESUMEN

A major challenge in current biology is to understand the genetic basis of variation for quantitative traits. We review the principles of quantitative trait locus mapping and summarize insights about the genetic architecture of quantitative traits that have been obtained over the past decades. We are currently in the midst of a genomic revolution, which enables us to incorporate genetic variation in transcript abundance and other intermediate molecular phenotypes into a quantitative trait locus mapping framework. This systems genetics approach enables us to understand the biology inside the 'black box' that lies between genotype and phenotype in terms of causal networks of interacting genes.


Asunto(s)
Ligamiento Genético , Carácter Cuantitativo Heredable , Animales , Mapeo Cromosómico , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA