Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
PLoS Pathog ; 12(6): e1005730, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27322179

RESUMEN

[This corrects the article DOI: 10.1371/journal.ppat.1005387.].

2.
PLoS Pathog ; 12(1): e1005387, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26799957

RESUMEN

Hosts encounter an ever-changing array of pathogens, so there is continual selection for novel ways to resist infection. A powerful way to understand how hosts evolve resistance is to identify the genes that cause variation in susceptibility to infection. Using high-resolution genetic mapping we have identified a naturally occurring polymorphism in a gene called Ge-1 that makes Drosophila melanogaster highly resistant to its natural pathogen Drosophila melanogaster sigma virus (DMelSV). By modifying the sequence of the gene in transgenic flies, we identified a 26 amino acid deletion in the serine-rich linker region of Ge-1 that is causing the resistance. Knocking down the expression of the susceptible allele leads to a decrease in viral titre in infected flies, indicating that Ge-1 is an existing restriction factor whose antiviral effects have been increased by the deletion. Ge-1 plays a central role in RNA degradation and the formation of processing bodies (P bodies). A key effector in antiviral immunity, the RNAi induced silencing complex (RISC), localises to P bodies, but we found that Ge-1-based resistance is not dependent on the small interfering RNA (siRNA) pathway. However, we found that Decapping protein 1 (DCP1) protects flies against sigma virus. This protein interacts with Ge-1 and commits mRNA for degradation by removing the 5' cap, suggesting that resistance may rely on this RNA degradation pathway. The serine-rich linker domain of Ge-1 has experienced strong selection during the evolution of Drosophila, suggesting that this gene may be under long-term selection by viruses. These findings demonstrate that studying naturally occurring polymorphisms that increase resistance to infections enables us to identify novel forms of antiviral defence, and support a pattern of major effect polymorphisms controlling resistance to viruses in Drosophila.


Asunto(s)
Proteínas Portadoras/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Polimorfismo Genético , Rhabdoviridae , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Drosophila melanogaster/virología , Genotipo , Datos de Secuencia Molecular , ARN Interferente Pequeño , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
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.
PLoS Genet ; 11(5): e1005163, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25933381

RESUMEN

Pigmentation varies within and between species and is often adaptive. The amount of pigmentation on the abdomen of Drosophila melanogaster is a relatively simple morphological trait, which serves as a model for mapping the genetic basis of variation in complex phenotypes. Here, we assessed natural variation in female abdominal pigmentation in 175 sequenced inbred lines of the Drosophila melanogaster Genetic Reference Panel, derived from the Raleigh, NC population. We quantified the proportion of melanization on the two most posterior abdominal segments, tergites 5 and 6 (T5, T6). We found significant genetic variation in the proportion of melanization and high broad-sense heritabilities for each tergite. Genome-wide association studies identified over 150 DNA variants associated with the proportion of melanization on T5 (84), T6 (34), and the difference between T5 and T6 (35). Several of the top variants associated with variation in pigmentation are in tan, ebony, and bric-a-brac1, genes known to affect D. melanogaster abdominal pigmentation. Mutational analyses and targeted RNAi-knockdown showed that 17 out of 28 (61%) novel candidate genes implicated by the genome-wide association study affected abdominal pigmentation. Several of these genes are involved in developmental and regulatory pathways, chitin production, cuticle structure, and vesicle formation and transport. These findings show that genetic variation may affect multiple steps in pathways involved in tergite development and melanization. Variation in these novel candidates may serve as targets for adaptive evolution and sexual selection in D. melanogaster.


Asunto(s)
Drosophila melanogaster/genética , Pigmentación/genética , Animales , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Evolución Molecular , Femenino , Estudios de Asociación Genética , Variación Genética , Desequilibrio de Ligamiento , Fenotipo , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
5.
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
6.
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
7.
Theor Appl Genet ; 129(12): 2413-2427, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27586153

RESUMEN

KEY MESSAGE: Predictive ability derived from gene expression and metabolic information was evaluated using genomic prediction methods based on datasets from a public maize panel. With the rapid development of high throughput biological technologies, information from gene expression and metabolites has received growing attention in plant genetics and breeding. In this study, we evaluated the utility of gene expression and metabolic information for genomic prediction using data obtained from a maize diversity panel. Our results show that, when used as predictor variables, gene expression levels and metabolite abundances provided reasonable predictive abilities relative to those based on genetic markers, although these values were not as large as those with genetic markers. Integrating gene expression levels and metabolite abundances with genetic markers significantly improved predictive abilities in comparison to the benchmark genomic best linear unbiased prediction model using genome-wide markers only. Predictive abilities based on gene expression and metabolites were trait-specific and were affected by the time of measurement and tissue samples as well as the number of genes and metabolites included in the model. In general, our results suggest that, rather than being conventionally used as intermediate phenotypes, gene expression and metabolic information can be used as predictors for genomic prediction and help improve genetic gains for complex traits in breeding programs.


Asunto(s)
Expresión Génica , Genoma de Planta , Genómica/métodos , Zea mays/genética , Marcadores Genéticos , Fenotipo , Fitomejoramiento
8.
Chem Senses ; 40(4): 233-43, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25687947

RESUMEN

The genetic underpinnings that contribute to variation in olfactory perception are not fully understood. To explore the genetic basis of variation in olfactory perception, we measured behavioral responses to 14 chemically diverse naturally occurring odorants in 260400 flies from 186 lines of the Drosophila melanogaster Genetic Reference Panel, a population of inbred wild-derived lines with sequenced genomes. We observed variation in olfactory behavior for all odorants. Low to moderate broad-sense heritabilities and the large number of tests for genotype-olfactory phenotype association performed precluded any individual variant from reaching formal significance. However, the top variants (nominal P < 5×10(-5)) were highly enriched for genes involved in nervous system development and function, as expected for a behavioral trait. Further, pathway enrichment analyses showed that genes tagged by the top variants included components of networks centered on cyclic guanosine monophosphate and inositol triphosphate signaling, growth factor signaling, Rho signaling, axon guidance, and regulation of neural connectivity. Functional validation with RNAi and mutations showed that 15 out of 17 genes tested indeed affect olfactory behavior. Our results show that in addition to chemoreceptors, variation in olfactory perception depends on polymorphisms that can result in subtle variations in synaptic connectivity within the nervous system.


Asunto(s)
Conducta Animal/fisiología , Drosophila melanogaster/genética , Drosophila melanogaster/fisiología , Variación Genética/genética , Percepción Olfatoria/genética , Percepción Olfatoria/fisiología , Animales
9.
PLoS Genet ; 8(11): e1003057, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23166512

RESUMEN

Variation in susceptibility to infectious disease often has a substantial genetic component in animal and plant populations. We have used genome-wide association studies (GWAS) in Drosophila melanogaster to identify the genetic basis of variation in susceptibility to viral infection. We found that there is substantially more genetic variation in susceptibility to two viruses that naturally infect D. melanogaster (DCV and DMelSV) than to two viruses isolated from other insects (FHV and DAffSV). Furthermore, this increased variation is caused by a small number of common polymorphisms that have a major effect on resistance and can individually explain up to 47% of the heritability in disease susceptibility. For two of these polymorphisms, it has previously been shown that they have been driven to a high frequency by natural selection. An advantage of GWAS in Drosophila is that the results can be confirmed experimentally. We verified that a gene called pastrel--which was previously not known to have an antiviral function--is associated with DCV-resistance by knocking down its expression by RNAi. Our data suggest that selection for resistance to infectious disease can increase genetic variation by increasing the frequency of major-effect alleles, and this has resulted in a simple genetic basis to variation in virus resistance.


Asunto(s)
Evolución Biológica , Resistencia a la Enfermedad/genética , Drosophila melanogaster , Estudio de Asociación del Genoma Completo , Alelos , Animales , Mapeo Cromosómico , Dicistroviridae/genética , Dicistroviridae/patogenicidad , Drosophila melanogaster/genética , Drosophila melanogaster/virología , Genotipo , Rhabdoviridae/genética , Rhabdoviridae/patogenicidad , Selección Genética
10.
PLoS Genet ; 8(12): e1003129, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23284297

RESUMEN

Wolbachia are maternally inherited symbiotic bacteria, commonly found in arthropods, which are able to manipulate the reproduction of their host in order to maximise their transmission. The evolutionary history of endosymbionts like Wolbachia can be revealed by integrating information on infection status in natural populations with patterns of sequence variation in Wolbachia and host mitochondrial genomes. Here we use whole-genome resequencing data from 290 lines of Drosophila melanogaster from North America, Europe, and Africa to predict Wolbachia infection status, estimate relative cytoplasmic genome copy number, and reconstruct Wolbachia and mitochondrial genome sequences. Overall, 63% of Drosophila strains were predicted to be infected with Wolbachia by our in silico analysis pipeline, which shows 99% concordance with infection status determined by diagnostic PCR. Complete Wolbachia and mitochondrial genomes show congruent phylogenies, consistent with strict vertical transmission through the maternal cytoplasm and imperfect transmission of Wolbachia. Bayesian phylogenetic analysis reveals that the most recent common ancestor of all Wolbachia and mitochondrial genomes in D. melanogaster dates to around 8,000 years ago. We find evidence for a recent global replacement of ancestral Wolbachia and mtDNA lineages, but our data suggest that the derived wMel lineage arose several thousand years ago, not in the 20th century as previously proposed. Our data also provide evidence that this global replacement event is incomplete and is likely to be one of several similar incomplete replacement events that have occurred since the out-of-Africa migration that allowed D. melanogaster to colonize worldwide habitats. This study provides a complete genomic analysis of the evolutionary mode and temporal dynamics of the D. melanogaster-Wolbachia symbiosis, as well as important resources for further analyses of the impact of Wolbachia on host biology.


Asunto(s)
Drosophila melanogaster , Metagenómica , Simbiosis , Wolbachia , Animales , Teorema de Bayes , Drosophila melanogaster/genética , Drosophila melanogaster/fisiología , Evolución Molecular , Variación Genética , Genoma Mitocondrial , Haplotipos , Filogenia , Wolbachia/genética , Wolbachia/fisiología
11.
Proc Natl Acad Sci U S A ; 109(39): 15553-9, 2012 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-22949659

RESUMEN

Epistasis-nonlinear genetic interactions between polymorphic loci-is the genetic basis of canalization and speciation, and epistatic interactions can be used to infer genetic networks affecting quantitative traits. However, the role that epistasis plays in the genetic architecture of quantitative traits is controversial. Here, we compared the genetic architecture of three Drosophila life history traits in the sequenced inbred lines of the Drosophila melanogaster Genetic Reference Panel (DGRP) and a large outbred, advanced intercross population derived from 40 DGRP lines (Flyland). We assessed allele frequency changes between pools of individuals at the extremes of the distribution for each trait in the Flyland population by deep DNA sequencing. The genetic architecture of all traits was highly polygenic in both analyses. Surprisingly, none of the SNPs associated with the traits in Flyland replicated in the DGRP and vice versa. However, the majority of these SNPs participated in at least one epistatic interaction in the DGRP. Despite apparent additive effects at largely distinct loci in the two populations, the epistatic interactions perturbed common, biologically plausible, and highly connected genetic networks. Our analysis underscores the importance of epistasis as a principal factor that determines variation for quantitative traits and provides a means to uncover genetic networks affecting these traits. Knowledge of epistatic networks will contribute to our understanding of the genetic basis of evolutionarily and clinically important traits and enhance predictive ability at an individualized level in medicine and agriculture.


Asunto(s)
Epistasis Genética/fisiología , Genes de Insecto/fisiología , Carácter Cuantitativo Heredable , Animales , Drosophila melanogaster , Polimorfismo de Nucleótido Simple
12.
Nat Genet ; 38(7): 824-9, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16783380

RESUMEN

The abundance of transposable elements and DNA repeat sequences in mammalian genomes raises the question of whether such insertions represent passive evolutionary baggage or may influence the expression of complex traits. We addressed this question in Drosophila melanogaster, in which the effects of single transposable elements on complex traits can be assessed in genetically identical individuals reared in controlled environments. Here we demonstrate that single P-element insertions in the intergenic region between the gustatory receptor 5a (Gr5a, also known as Tre) and trapped in endoderm 1 (Tre1), which encodes an orphan receptor, exert complex pleiotropic effects on fitness traits, including selective nutrient intake, life span, and resistance to starvation and heat stress. Mutations in this region interact epistatically with downstream components of the insulin signaling pathway. Transposon-induced sex-specific and sex-antagonistic effects further accentuate the complex influences that intergenic transposable elements can contribute to quantitative trait phenotypes.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Receptores de Superficie Celular/genética , Receptores Acoplados a Proteínas G/genética , Animales , Elementos Transponibles de ADN/genética , ADN Intergénico/genética , Drosophila melanogaster/fisiología , Epistasis Genética , Femenino , Genes de Insecto , Insulina/metabolismo , Longevidad/genética , Masculino , Mutación , Fenotipo , Carácter Cuantitativo Heredable , Secuencias Repetitivas de Ácidos Nucleicos , Transducción de Señal , Gusto/genética
13.
PLoS Genet ; 7(10): e1002337, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22028673

RESUMEN

To understand the molecular basis of how hosts evolve resistance to their parasites, we have investigated the genes that cause variation in the susceptibility of Drosophila melanogaster to viral infection. Using a host-specific pathogen of D. melanogaster called the sigma virus (Rhabdoviridae), we mapped a major-effect polymorphism to a region containing two paralogous genes called CHKov1 and CHKov2. In a panel of inbred fly lines, we found that a transposable element insertion in the protein coding sequence of CHKov1 is associated with increased resistance to infection. Previous research has shown that this insertion results in a truncated messenger RNA that encodes a far shorter protein than the susceptible allele. This resistant allele has rapidly increased in frequency under directional selection and is now the commonest form of the gene in natural populations. Using genetic mapping and site-specific recombination, we identified a third genotype with considerably greater resistance that is currently rare in the wild. In these flies there have been two duplications, resulting in three copies of both the truncated allele of CHKov1 and CHKov2 (one of which is also truncated). Remarkably, the truncated allele of CHKov1 has previously been found to confer resistance to organophosphate insecticides. As estimates of the age of this allele predate the use of insecticides, it is likely that this allele initially functioned as a defence against viruses and fortuitously "pre-adapted" flies to insecticides. These results demonstrate that strong selection by parasites for increased host resistance can result in major genetic changes and rapid shifts in allele frequencies; and, contrary to the prevailing view that resistance to pathogens can be a costly trait to evolve, the pleiotropic effects of these changes can have unexpected benefits.


Asunto(s)
Elementos Transponibles de ADN/genética , Resistencia a la Enfermedad/genética , Drosophila melanogaster/genética , Drosophila melanogaster/virología , Rhabdoviridae , Duplicaciones Segmentarias en el Genoma/genética , Alelos , Animales , Evolución Biológica , Mapeo Cromosómico , Frecuencia de los Genes , Variación Genética , Mutagénesis Insercional/genética , ARN Mensajero/genética , Selección Genética
14.
Proc Natl Acad Sci U S A ; 108(41): 17070-5, 2011 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-21949384

RESUMEN

Epistasis and pleiotropy feature prominently in the genetic architecture of quantitative traits but are difficult to assess in outbred populations. We performed a diallel cross among coisogenic Drosophila P-element mutations associated with hyperaggressive behavior and showed extensive epistatic and pleiotropic effects on aggression, brain morphology, and genome-wide transcript abundance in head tissues. Epistatic interactions were often of greater magnitude than homozygous effects, and the topology of epistatic networks varied among these phenotypes. The transcriptional signatures of homozygous and double heterozygous genotypes derived from the six mutations imply a large mutational target for aggressive behavior and point to evolutionarily conserved genetic mechanisms and neural signaling pathways affecting this universal fitness trait.


Asunto(s)
Agresión/fisiología , Conducta Animal/fisiología , Drosophila melanogaster/genética , Drosophila melanogaster/fisiología , Animales , Encéfalo/anatomía & histología , Cruzamientos Genéticos , Drosophila melanogaster/anatomía & histología , Epistasis Genética , Evolución Molecular , Femenino , Expresión Génica , Redes Reguladoras de Genes , Genes de Insecto , Masculino , Mutación , Fenotipo
15.
PLoS Genet ; 6(7): e1001037, 2010 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-20686706

RESUMEN

Understanding the genetic and environmental factors that affect variation in life span and senescence is of major interest for human health and evolutionary biology. Multiple mechanisms affect longevity, many of which are conserved across species, but the genetic networks underlying each mechanism and cross-talk between networks are unknown. We report the results of a screen for mutations affecting Drosophila life span. One third of the 1,332 homozygous P-element insertion lines assessed had quantitative effects on life span; mutations reducing life span were twice as common as mutations increasing life span. We confirmed 58 mutations with increased longevity, only one of which is in a gene previously associated with life span. The effects of the mutations increasing life span were highly sex-specific, with a trend towards opposite effects in males and females. Mutations in the same gene were associated with both increased and decreased life span, depending on the location and orientation of the P-element insertion, and genetic background. We observed substantial--and sex-specific--epistasis among a sample of ten mutations with increased life span. All mutations increasing life span had at least one deleterious pleiotropic effect on stress resistance or general health, with different patterns of pleiotropy for males and females. Whole-genome transcript profiles of seven of the mutant lines and the wild type revealed 4,488 differentially expressed transcripts, 553 of which were common to four or more of the mutant lines, which include genes previously associated with life span and novel genes implicated by this study. Therefore longevity has a large mutational target size; genes affecting life span have variable allelic effects; alleles affecting life span exhibit antagonistic pleiotropy and form epistatic networks; and sex-specific mutational effects are ubiquitous. Comparison of transcript profiles of long-lived mutations and the control line reveals a transcriptional signature of increased life span.


Asunto(s)
Drosophila/genética , Longevidad/genética , Mutación , Envejecimiento/genética , Animales , Evolución Biológica , Epigénesis Genética , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Factores Sexuales
16.
G3 (Bethesda) ; 11(10)2021 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-34568933

RESUMEN

Epistasis-gene-gene interaction-is common for mutations with large phenotypic effects in humans and model organisms. Epistasis impacts quantitative genetic models of speciation, response to natural and artificial selection, genetic mapping, and personalized medicine. However, the existence and magnitude of epistasis between alleles with small quantitative phenotypic effects are controversial and difficult to assess. Here, we use the Drosophila melanogaster Genetic Reference Panel of sequenced inbred lines to evaluate the magnitude of naturally occurring epistasis modifying the effects of mutations in jing and inv, two transcription factors that have subtle quantitative effects on head morphology as homozygotes. We find significant epistasis for both mutations and performed single marker genome-wide association analyses to map candidate modifier variants and loci affecting head morphology. A subset of these loci was significantly enriched for a known genetic interaction network, and mutations of the candidate epistatic modifier loci also affect head morphology.


Asunto(s)
Drosophila melanogaster , Epistasis Genética , Alelos , Animales , Mapeo Cromosómico , Drosophila melanogaster/genética , Estudio de Asociación del Genoma Completo
17.
Elife ; 52016 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-27213517

RESUMEN

Mutation and natural selection shape the genetic variation in natural populations. Here, we directly estimated the spontaneous mutation rate by sequencing new Drosophila mutation accumulation lines maintained with minimal natural selection. We inferred strong stabilizing natural selection on quantitative traits because genetic variation among wild-derived inbred lines was much lower than predicted from a neutral model and the mutational effects were much larger than allelic effects of standing polymorphisms. Stabilizing selection could act directly on the traits, or indirectly from pleiotropic effects on fitness. However, our data are not consistent with simple models of mutation-stabilizing selection balance; therefore, further empirical work is needed to assess the balance of evolutionary forces responsible for quantitative genetic variation.


Asunto(s)
Drosophila/genética , Variación Genética , Tasa de Mutación , Animales , Acumulación de Mutaciones , Sitios de Carácter Cuantitativo , Selección Genética
18.
PLoS One ; 10(5): e0126880, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25950439

RESUMEN

The ability to predict quantitative trait phenotypes from molecular polymorphism data will revolutionize evolutionary biology, medicine and human biology, and animal and plant breeding. Efforts to map quantitative trait loci have yielded novel insights into the biology of quantitative traits, but the combination of individually significant quantitative trait loci typically has low predictive ability. Utilizing all segregating variants can give good predictive ability in plant and animal breeding populations, but gives little insight into trait biology. Here, we used the Drosophila Genetic Reference Panel to perform both a genome wide association analysis and genomic prediction for the fitness-related trait chill coma recovery time. We found substantial total genetic variation for chill coma recovery time, with a genetic architecture that differs between males and females, a small number of molecular variants with large main effects, and evidence for epistasis. Although the top additive variants explained 36% (17%) of the genetic variance among lines in females (males), the predictive ability using genomic best linear unbiased prediction and a relationship matrix using all common segregating variants was very low for females and zero for males. We hypothesized that the low predictive ability was due to the mismatch between the infinitesimal genetic architecture assumed by the genomic best linear unbiased prediction model and the true genetic architecture of chill coma recovery time. Indeed, we found that the predictive ability of the genomic best linear unbiased prediction model is markedly improved when we combine quantitative trait locus mapping with genomic prediction by only including the top variants associated with main and epistatic effects in the relationship matrix. This trait-associated prediction approach has the advantage that it yields biologically interpretable prediction models.


Asunto(s)
Drosophila/genética , Drosophila/fisiología , Genes de Insecto , Animales
19.
PLoS One ; 10(7): e0132980, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26167869

RESUMEN

[This corrects the article DOI: 10.1371/journal.pone.0126880.].

20.
Nat Commun ; 6: 10115, 2015 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-26656654

RESUMEN

Genetic variation in brain size may provide the basis for the evolution of the brain and complex behaviours. The genetic substrate and the selective pressures acting on brain size are poorly understood. Here we use the Drosophila Genetic Reference Panel to map polymorphic variants affecting natural variation in mushroom body morphology. We identify 139 genes and 39 transcription factors and confirm effects on development and adult plasticity. We show correlations between morphology and aggression, sleep and lifespan. We propose that natural variation in adult brain size is controlled by interaction of the environment with gene networks controlling development and plasticity.


Asunto(s)
Drosophila melanogaster/anatomía & histología , Drosophila melanogaster/genética , Cuerpos Pedunculados/anatomía & histología , Animales , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Regulación de la Expresión Génica/fisiología , Masculino , Interferencia de ARN , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA