首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 62 毫秒
1.
Insertional mutagenesis screens play an integral part in the annotating of functional data for all sequenced genes in the postgenomic era. Chemical mutagenesis screens are highly efficient but identifying the causative gene can be a laborious task. Other mutagenesis platforms, such as transposable elements, have been successfully applied for insertional mutagenesis screens in both the mouse and rat. However, relatively low transposition efficiency has hampered their use as a high-throughput forward genetic mutagenesis screen. Here we report the first evidence of germline activity in the mouse using a naturally active DNA transposon derived from the medaka fish called Tol2, as an alternative system for high-throughput forward genetic mutagenesis screening tool.THE Tol2 (transposable element of Oryzias latipes, number 2) element belongs to the hAT family (hobo of Drosophilia, Activator of maize and Tam3 of snapdragon) of transposons and was the first known autonomously active vertebrate type II transposable element (Koga et al. 1996; Kawakami et al. 1998). Unlike other DNA-type transposons like Sleeping Beauty (SB) (Ivics et al. 1997) or piggyBac (PB) (Fraser et al. 1996), Tol2 does not exhibit any known strong site specificity for integration nor does it exhibit any significant overexpression inhibition activity (Kawakami and Noda 2004; Balciunas et al. 2006) as seen in SB (Geurts et al. 2003). Recently, Tol2 was shown to effectively carry large DNA cargo of up to 10 kb in human and mouse cells without affecting its transposition efficiency (Balciunas et al. 2006). To date, Tol2 has also been demonstrated to transpose efficiently in zebrafish, frog, chicken, mouse cells, and human cells (Kawakami et al. 2000, 2004; Koga et al. 2003; Kawakami and Noda 2004; Balciunas et al. 2006; Hamlet et al. 2006; Sato et al. 2007).Germline mutagenesis using the SB transposon system has been demonstrated in both the mouse (Dupuy et al. 2001; Horie et al. 2001) and rat (Kitada et al. 2007; Lu et al. 2007). In addition, PB germline mutagenesis in mice has also been demonstrated (Ding et al. 2005; Wu et al. 2007). However, the relatively low germline transposition efficiency of both transposon systems reported so far has hampered their use in a high-throughput forward genetic mutagenesis screen (Keng et al. 2005; Kitada et al. 2007).

TABLE 1

Germline transposition frequency in various transposon systems
Transposon systemAverage transposition events per gameteMouse strainReference
SB2FVB/NDupuy et al. (2001)
SB1.25C3H and C57BL/6Horie et al. (2001)
SB1.15C3H and C57BL/6Keng et al. (2005)
PB1.1FVB/NDing et al. (2005)
PB1C57BL/6Wu et al. (2007)
Tol2
3
FVB/N
Present study
Open in a separate windowSB, Sleeping Beauty; PB, piggyBac; Tol2, transposable element of Oryzias latipes, number 2.In search of an alternative tool for high-throughput forward germline mutagenesis screen in mice, a Tol2 transposon insertional mutagenesis system was generated on the basis of a similar strategy used for the SB transposon system (Horie et al. 2003; Keng et al. 2005). In the present study, we successfully demonstrate the novel use of the Tol2 transposon system for germline mutagenesis in mouse. Our results indicate the potential use of this transposon system for a high-throughput, large-scale forward mutagenesis screen in the mouse germline.  相似文献   

2.
3.
4.
5.
The intraflagellar transport machinery is required for the assembly of cilia. It has been investigated by biochemical, genetic, and computational methods that have identified at least 21 proteins that assemble into two subcomplexes. It has been hypothesized that complex A is required for retrograde transport. Temperature-sensitive mutations in FLA15 and FLA17 show defects in retrograde intraflagellar transport (IFT) in Chlamydomonas. We show that IFT144 and IFT139, two complex A proteins, are encoded by FLA15 and FLA17, respectively. The fla15 allele is a missense mutation in a conserved cysteine and the fla17 allele is an in-frame deletion of three exons. The flagellar assembly defect of each mutant is rescued by the respective transgenes. In fla15 and fla17 mutants, bulges form in the distal one-third of the flagella at the permissive temperature and this phenotype is also rescued by the transgenes. These bulges contain the complex B component IFT74/72, but not α-tubulin or p28, a component of an inner dynein arm, which suggests specificity with respect to the proteins that accumulate in these bulges. IFT144 and IFT139 are likely to interact with each other and other proteins on the basis of three distinct genetic tests: (1) Double mutants display synthetic flagellar assembly defects at the permissive temperature, (2) heterozygous diploid strains exhibit second-site noncomplemention, and (3) transgenes confer two-copy suppression. Since these tests show different levels of phenotypic sensitivity, we propose they illustrate different gradations of gene interaction between complex A proteins themselves and with a complex B protein (IFT172).CILIA and flagella are microtubule-based organelles that are found on most mammalian cells. They provide motility to cells and participate in many sensory processes. Defects in or loss of cilia/flagella cause a variety of human diseases that include polycystic kidney disease, retinal degeneration, infertility, obesity, respiratory defects, left–right axis determination, and polydactyly (Fliegauf et al. 2007). Mouse mutants demonstrate that cilia are essential for viability, neural tube closure, and bone development (Eggenschwiler and Anderson 2007; Fliegauf et al. 2007). Cilia and flagella are also present in protists, algae, moss, and some fungi.The assembly and maintenance of cilia and flagella require intraflagellar transport (IFT) (Kozminski et al. 1995). IFT involves the movement of 100- to 200-nm-long protein particles from the basal body located in the cell body to the tip of the flagella using the heterotrimeric kinesin-2 (anterograde movement) (Kozminski et al. 1995) and movement back to the cell body (retrograde movement) using the cytoplasmic dynein complex (Pazour et al. 1999; Porter et al. 1999). IFT particles change their direction of movement as well as their size, speed, and frequency at the ends of the flagella as they switch from anterograde to retrograde movement (Iomini et al. 2001). Biochemical isolation of IFT particles reveals that they are composed of at least 16 proteins and that these particles can be dissociated into two complexes in vitro by changing the salt concentration (Cole et al. 1998; Piperno et al. 1998). Recent genetic and bioinformatics analysis adds at least 7 more proteins to the IFT particle (Follit et al. 2009) (Eggenschwiler and Anderson 2007).

TABLE 1

Proteins and gene names for the intraflagellar transport particles in Chlamydomonas, C. elegans, and mouse
ProteinMotifChlamydomonas geneC. elegans geneMouse geneReferences to worm and mouse genes
Complex A
IFT144WDFLA15
IFT140WDche-11Qin et al. (2001)
IFT139TRPFLA17dyf-2THM1Efimenko et al. (2006); Tran et al. (2008)
IFT122WDIFTA-1Blacque et al. (2006)
IFT121WDdaf-10Bell et al. (2006)
IFT43
Complex B
IFT172WDFLA11osm-1WimpleHuangfu et al. (2003); Pedersen et al. (2005); Bell et al. (2006)
IFT88TRPIFT88osm-5Tg737/PolarisPazour et al. (2000); Qin et al. (2001)
IFT81Coilift-81CDV1Kobayashi et al. (2007)
IFT80WDche-2Wdr56Fujiwara et al. (1999)
IFT74/72Coilift-74Cmg1Kobayashi et al. (2007)
IFT57/55Coilche-13HippiHaycraft et al. (2003)
IFT54Microtubule binding domain MIP-T3dyf-11Traf3IP1Kunitomo and Iino (2008); Li et al. (2008); Omori et al. (2008); Follit et al. (2009)
IFT52ABC typeBLD1osm-6Ngd2Brazelton et al. (2001); Bell et al. (2006)
IFT46IFT46dyf-6Bell et al. (2006); Hou et al. (2007)
IFT27G proteinNot presentRabl4
IFT25Hsp20Not presentHSP16.1Follit et al. (2009)
IFT22G proteinIFTA-2Rabl5Schafer et al. (2006)
IFT20CoilFollit et al. (2006)
FAP22Cluamp related proteindyf-3Cluamp1Murayama et al. (2005); Follit et al. (2009)
DYF13


dyf-13
Ttc26
Blacque et al. (2005)
Open in a separate window—, no mutant found to date in Chlamydomonas.A collection of temperature-sensitive mutant strains that fail to assemble flagella at the restrictive temperature of 32° was isolated in Chlamydomonas (Huang et al. 1977; Adams et al. 1982; Piperno et al. 1998; Iomini et al. 2001). Analysis of the flagella at 21° permits the measurement of the velocity and frequency of IFT particles in the mutant strains. This analysis suggested that assembly has four phases: recruitment to the basal body, anterograde movement (phases I and II), retrograde movement, and return to the cytoplasm (phases III and IV) (Iomini et al. 2001). Different mutants were classified as defective in these four phases. However, because different alleles of FLA8 were classified as defective in different phases (Iomini et al. 2001; Miller et al. 2005), we combined mutants with IFT defects into just two classes. The first group (phases I and II) includes mutant strains that show decreased anterograde velocities, a decreased ratio of anterograde to retrograde particles, and an accumulation of complex A proteins at the basal body. This group includes mutations in the FLA8 and FLA10 genes, which encode the two motor subunits of kinesin-2 (Walther et al. 1994; Miller et al. 2005), as well as mutations in three unknown genes (FLA18, FLA27, and FLA28). The second group includes mutant strains that show the reciprocal phenotype (phases III and IV); these phenotypes include decreased retrograde velocities, an increased ratio of anterograde to retrograde particles, and an accumulation of complex B proteins in the flagella. With the exception of the FLA11 gene, which encodes IFT172, a component of complex B (Pedersen et al. 2005), the gene products in this class are unknown (FLA2, FLA15, FLA16, FLA17, and FLA24). One might predict that mutations in this group would map to genes that encode complex A or retrograde motor subunits. Interestingly, IFT particles isolated from fla11, fla15, fla16, and fla17-1 flagella show depletion of complex A polypeptides (Piperno et al. 1998; Iomini et al. 2001). The inclusion of IFT172 in this class is explained by the observations that IFT172 plays a role in remodeling the IFT particles at the flagellar tip to transition from anterograde to retrograde movement (Pedersen et al. 2005). The remaining mutant strains do not show obvious defects in velocities, ratios, or accumulation at 21° and may reflect a less severe phenotype at the permissive temperature or a non-IFT role for these genes.Direct interactions occur between components of complex B. IFT81 and IFT74/72 interact to form a scaffold required for IFT complex B assembly (Lucker et al. 2005). IFT57 and IFT20 also interact with each other and kinesin-2 (Baker et al. 2003). While physical interactions are being used to define IFT particle architecture, genetic interactions among loci encoding IFT components should be instructive regarding their function as well. To probe retrograde movement and its function, we have identified the gene products encoded by two retrograde defective mutant strains. They are FLA15 and FLA17 and encode IFT144 and IFT139, respectively. The genetic interactions of these loci provide interesting clues about the assembly of the IFT particles and possible physical interactions in the IFT particles.  相似文献   

6.
We used a large panel of pedigreed, genetically admixed house mice to study patterns of recombination rate variation in a leading mammalian model system. We found considerable inter-individual differences in genomic recombination rates and documented a significant heritable component to this variation. These findings point to clear variation in recombination rate among common laboratory strains, a result that carries important implications for genetic analysis in the house mouse.THE rate of recombination—the amount of crossing over per unit DNA—is a key parameter governing the fidelity of meiosis. Recombination rates that are too high or too low frequently give rise to aneuploid gametes or prematurely arrest the meiotic cell cycle (Hassold and Hunt 2001). As a consequence, recombination rates should experience strong selective pressures to lie within the range defined by the demands of meiosis (Coop and Przeworski 2007). Nonetheless, classical genetic studies in Drosophila (Chinnici 1971; Kidwell 1972; Brooks and Marks 1986), crickets (Shaw 1972), flour beetles (Dewees 1975), and lima beans (Allard 1963) have shown that considerable inter-individual variation for recombination rate is present within populations. Recent studies examining the transmission of haplotypes in human pedigrees have corroborated these findings (Broman et al. 1998; Kong et al. 2002; Coop et al. 2008).Here, we use a large panel of heterogeneous stock (HS) mice to study variation in genomic recombination rates in a genetic model system. These mice are genetically admixed, derived from an initial generation of pseudorandom mating among eight common inbred laboratory strains (DBA/2J, C3H/HeJ, AKR/J, A/J, BALB/cJ, CBA/J, C57BL/6J, and LP/J), followed by >50 generations of pseudorandom mating in subsequent hybrid cohorts (Mott et al. 2000; Demarest et al. 2001). The familial relationships among animals in recent generations were tracked to organize the mice into pedigrees. In total, this HS panel includes ∼2300 animals comprising 85 families, 8 of which span multiple generations. The remainder consists of nuclear families (sibships) that range from 1 to 34 sibs, with an average of 9.6 sibs (Valdar et al. 2006) (Mott et al. 2000; Demarest et al. 2001; Shifman et al. 2006).

TABLE 1

Heterogeneous stock mouse pedigrees
PedigreePedigree classNo. of nonoverlapping sibships in the pedigreeNo. of retained sibshipsNo. of meioses
1Multigenerational1717464
2Multigenerational2720728
3Multigenerational2319602
4Multigenerational149254
5Multigenerational119242
6Multigenerational5368
7Multigenerational43100
8Multigenerational2116
9Sibshipa2120
32–85Sibship511146
Total1801323640
Open in a separate windowaThis family was composed of two sibships sharing a common mother but with different fathers.With the exception of several founding individuals, most of these HS mice have been genotyped at 13,367 single nucleotide polymorphisms (SNPs) across the genome (available at http://gscan.well.ox.ac.uk/). Although the publicly available HS genotypes have passed data quality filters (Shifman et al. 2006), we took several additional measures to ensure the highest possible accuracy of base calls. First, data were cleansed of all non-Mendelian inheritances, and genotypes with quality scores <0.4 were removed. Genotypes that resulted in tight (<10 cM in sex-specific distance) double recombinants were also omitted because strong positive crossover interference in the mouse renders such closely spaced crossovers biologically very unlikely (Broman et al. 2002). A total of 10,195 SNPs (including 298 on the X chromosome) passed these additional quality control criteria; the results presented below consider only this subset of highly accurate (>99.98%) and complete (<0.01% missing) genotypes. The cleaned data are publicly available (at http://cgd.jax.org/mousemapconverter/).We used the chrompic program within CRI-MAP (Lander and Green 1987; Green et al. 1990) to estimate the number of recombination events in parental meioses. The algorithm implemented in chrompic first phases parent and offspring genotypes using a maximum-likelihood approach. Next, recombination events occurring in the parental germline are identified by comparing parent and offspring haplotypes across the genome (Green et al. 1990). For example, a haplotype that first copies from one maternal chromosome and then switches to copying from the other maternal chromosome signals a recombination event in the maternal germline.chrompic is very memory intensive and cannot handle the multigenerational pedigrees and the large sibships included in the HS panel. To circumvent these computational limitations, several modifications to the data were implemented. First, the eight multigenerational pedigrees were split into 102 nonoverlapping sibships, retaining grandparental information when available (Cox et al. 2009). Finally, large sibships were subdivided: sibships with >13 progeny were split into two groups: those with >26 progeny were split into three groups and those with >39 sibs were split into four groups. Partitioning large sibships by units of 10, 11, or 12, rather than 13, had no effect on the estimation of crossover counts, suggesting that the estimates were robust to the unit of subdivision. These subdivided families were used only for haplotype inference; all other analyses treated whole sibships as focal units. In total, we analyzed 132 nonoverlapping sibships, ranging in size from 2 to 48 sibs (mean = 13.9). This data set encompassed 3640 meioses—300–2000% more meioses than previously studied human pedigrees (Broman et al. 1998; Kong et al. 2002; Coop et al. 2008)—providing excellent power to detect recombination rate variation among individuals.The recombination rate for the maternal (or paternal) parent of a given sibship was estimated as the average number of recombination events in the haploid maternal (or paternal) genomes transmitted to her (or his) offspring. Our analyses treat males and females separately, as previous observations in mice (Murray and Snell 1945; Mallyon 1951; Reeves et al. 1990; Dietrich et al. 1996; Shifman et al. 2006; Paigen et al. 2008), along with findings from this study, point to systematically higher recombination rates in female than in male mice (this study: P < 2.2 × 10−16, Mann–Whitney U-Test comparing autosomal crossover counts in the 131 HS females to those in the 131 HS males).There is considerable recombination rate heterogeneity among the 131 mothers and 131 fathers in the HS pedigrees (Figure 1). The female with the highest recombination rate had an average of nearly twice as many crossovers per meiosis compared with the lowest (female range: 9.0–17.3; mean = 13.3; SD = 3.28). Similarly, the least actively recombining male had only 55% the amount of recombination as the male with the highest recombination rate (male range: 7.7–14.7; mean = 11.7; SD = 2.76). These average values are similar to previously reported recombination counts in house mice, determined using both cytological (Dumas and Britton-Davidian 2002; Koehler et al. 2002) and genetic (Dietrich et al. 1996) approaches. Note that the recombination rates that we report reflect the number of exchange events visible in genetic data. Under the assumption of no chromatid interference, the expected number of crossovers that occur at meiosis is equal to twice these values.Open in a separate windowFigure 1.—Variation in recombination frequency in HS mice. The mean number of recombination events per transmitted gamete in each mother (A; n = 131) and father (B; n = 131) was inferred by comparing parent and offspring genotypes at >10,000 autosomal and X-linked markers using the CRIMAP chrompic computer program. Error bars span ±2 SEs.To test for variation in recombination within the HS females and within the HS males, we performed a one-way ANOVA using parental identity as the factor and the recombination count for a single haploid genome transmission on the pedigree as the response variable. Significance of the resultant F-statistic was empirically assessed by randomizing parental identity with respect to individual recombination counts, recomputing the F-statistic on the permuted data set, and determining the quantile position of the observed F-statistic along the distribution of 106 F-statistics derived from randomization. There is highly significant variation for genomic recombination rate among HS females (F = 1.7842, P < 10−6; Figure 1A) and males (F = 2.3103, P < 10−6; Figure 1B).We next examined patterns of recombination rate inheritance using the eight complex families to test for heritability of this trait. We fit a polygenic model of inheritance using the polygenic command within SOLAR v.4, accounting for the uneven relatedness among individuals through a matrix of pairwise coefficients of relatedness (Almasy and Blangero 1998). Sex was included as a covariate in the model to account for the well-established differences between male and female recombination rates in mice (Murray and Snell 1945; Mallyon 1951; Reeves et al. 1990; Dietrich et al. 1996; Shifman et al. 2006; Paigen et al. 2008). Recombination rates show significant narrow-sense heritability (h2 = 0.46; SE = 0.20; P = 0.008), indicating that variation for recombination rate among HS mice is partly attributable to additive genetic variation. This result agrees with previous evidence for genetic effects on recombination rate variation in the house mouse (Reeves et al. 1990; Shiroishi et al. 1991; Koehler et al. 2002).In summary, we have shown that HS mice differ significantly in their genomic recombination rates and have demonstrated that this variation is heritable. These findings indicate that interstrain variation for genomic average recombination rate exists among at least two of the eight progenitor strains of the HS stock, mirroring observations of significant variation among inbred laboratory strains for many other quantitative characters (Grubb et al. 2009). Indeed, cytological analyses have already revealed significant differences in recombination frequencies between A/J and C57BL/6J males (Koehler et al. 2002), two of the HS founding strains.This interstrain variation in genomic recombination rate carries important practical implications for genetic analysis in the house mouse. Most notably, crosses using inbred mouse strains with high recombination rates will provide higher mapping resolution than crosses using strains with reduced recombination rates. However, the strategic use of high-recombination-rate strains will not necessarily expedite the fine mapping of loci. The distribution of recombination events in mice is not uniform across chromosomes and appears to be strain specific (Paigen et al. 2008; Grey et al. 2009; Parvanov et al. 2009).The history of the classical inbred mouse strains as inferred from pedigrees (Beck et al. 2000), sequence comparisons to wild mice (Salcedo et al. 2007), and genomewide phylogenetic analyses (Frazer et al. 2007; Yang et al. 2007) suggests that much of the interstrain variation for recombination rate arises from genetic polymorphism among Mus domesticus individuals in nature. However, many other factors have likely shaped recombination rate variation among the classical strains, including inbreeding, artificial selection, and hybridization with closely related species (Wade and Daly 2005). These aspects of the laboratory mouse''s history challenge comparisons between recombination rate variation in the HS panel and human populations and provide strong motivation for studies of recombination rate variation in natural populations of house mice.Although we find a strong genetic component to inter-individual variation in recombination rate, a large fraction (∼54%) of the phenotypic variation for recombination is not explained by additive genetic variation alone. Sampling error and other forms of genetic variation (e.g., dominance and epistasis) likely combine to account for some of the residual variation. In addition, micro-environmental differences within the laboratory setting (Koren et al. 2002) and life history differences among families, including parental age (Koehler et al. 2002; Kong et al. 2004), might contribute to variation in recombination rates among the HS mice.Identifying the genetic loci that underlie recombination rate differences among the HS mice (and hence in the eight founding inbred strains) presents a logical next step in the research program initiated here. The complicated pedigree structure, relatively small number of animals with recombination rate estimates (n = 262), and potentially sex-specific genetic architecture of this trait (Kong et al. 2008; Paigen et al. 2008) will pose challenges to this analysis. Nonetheless, dissecting the genetic basis of recombination rate variation is a pursuit motivated by its potential to lend key insights into several enduring questions. Why do males and females differ in the rate and distribution of crossover events? What are the evolutionary mechanisms that give rise to intraspecific polymorphism and interspecific divergence for recombination rate? What are the functional consequences of recombination rate variation? Alternative experimental approaches, including those that combine the power of QTL mapping with immunocytological assays for measuring recombination rates in situ (Anderson et al. 1999), promise to offer additional clues onto the genetic mechanisms that give rise to variation in this important trait.  相似文献   

7.
Modern genomewide association studies are characterized by the problem of “missing heritability.” Epistasis, or genetic interaction, has been suggested as a possible explanation for the relatively small contribution of single significant associations to the fraction of variance explained. Of particular concern to investigators of genetic interactions is how to best represent and define epistasis. Previous studies have found that the use of different quantitative definitions for genetic interaction can lead to different conclusions when constructing genetic interaction networks and when addressing evolutionary questions. We suggest that instead, multiple representations of epistasis, or epistatic “subtypes,” may be valid within a given system. Selecting among these epistatic subtypes may provide additional insight into the biological and functional relationships among pairs of genes. In this study, we propose maximum-likelihood and model selection methods in a hypothesis-testing framework to choose epistatic subtypes that best represent functional relationships for pairs of genes on the basis of fitness data from both single and double mutants in haploid systems. We gauge the performance of our method with extensive simulations under various interaction scenarios. Our approach performs reasonably well in detecting the most likely epistatic subtype for pairs of genes, as well as in reducing bias when estimating the epistatic parameter (ɛ). We apply our approach to two available data sets from yeast (Saccharomyces cerevisiae) and demonstrate through overlap of our identified epistatic pairs with experimentally verified interactions and functional links that our results are likely of biological significance in understanding interaction mechanisms. We anticipate that our method will improve detection of epistatic interactions and will help to unravel the mysteries of complex biological systems.UNDERSTANDING the nature of genetic interactions is crucial to obtaining a more complete picture of complex biological systems and their evolution. The discovery of genetic interactions has been the goal of many researchers studying a number of model systems, including but not limited to Saccharomyces cerevisiae, Caenorhabditis elegans, and Escherichia coli (You and Yin 2002; Burch et al. 2003; Burch and Chao 2004; Tong et al. 2004; Drees et al. 2005; Sanjuán et al. 2005; Segre et al. 2005; Pan et al. 2006; Zhong and Sternberg 2006; Jasnos and Korona 2007; St. Onge et al. 2007; Decourty et al. 2008). Recently, high-throughput experimental approaches, such as epistatic mini-array profiles (E-MAPs) and genetic interaction analysis technology for E. coli (GIANT-coli), have enabled the study of epistasis on a large scale (Schuldiner et al. 2005, 2006; Collins et al. 2006, 2007; Typas et al. 2008). However, it remains unclear whether the computational and statistical methods currently in use to identify these interactions are indeed the most appropriate.The study of genetic interaction, or “epistasis,” has had a long and somewhat convoluted history. Bateson (1909) first used the term epistasis to describe the ability of a gene at one locus to “mask” the mutational influence of a gene at another locus (Cordell 2002). The term “epistacy” was later coined by Fisher (1918) to denote the statistical deviation of multilocus genotype values from an additive linear model for the value of a phenotype (Phillips 1998, 2008).These origins are the basis for the two main current interpretations of epistasis. The first, as introduced by Bateson (1909), is the “biological,” “physiological,” or “compositional” form of epistasis, concerned with the influence of an individual''s genetic background on an allele''s effect on phenotype (Cheverud and Routman 1995; Phillips 1998, 2008; Cordell 2002; Moore and Williams 2005). The second interpretation, attributed to Fisher, is “statistical” epistasis, which in its linear regression framework places the phenomenon of epistasis in the context of a population (Wagner et al. 1998; Wade et al. 2001; Wilke and Adami 2001; Moore and Williams 2005; Phillips 2008). Each of these approaches is equally valid in studying genetic interactions; however, confusion still exists about how to best reconcile the methods and results of the two (Phillips 1998, 2008; Cordell 2002; Moore and Williams 2005; Liberman and Feldman 2006; Aylor and Zeng 2008).Aside from the distinction between the statistical and the physiological definitions of epistasis, inconsistencies exist when studying solely physiological epistasis. For categorical traits, physiological epistasis is clear as a “masking” effect. When noncategorical or numerical traits are measured, epistasis is defined as the deviation of the phenotype of the multiple mutant from that expected under independence of the underlying genes.The “expectation” of the phenotype under independence, that is, in the absence of epistasis, is not defined consistently between studies. For clarity, consider epistasis between pairs of genes and, without loss of generality, consider fitness as the phenotype. The first commonly used definition of independence, originating from additivity, defines the effect of two independent mutations to be equal to the sum of the individual mutational effects. A second, motivated by the use of fitness as a phenotype, defines the effect of the two mutations as the product of the individual effects (Elena and Lenski 1997; Desai et al. 2007; Phillips 2008). A third definition of independence has been referred to as “minimum,” where alleles at two loci are independent if the double mutant has the same fitness as the less-fit single mutant. Mani et al. (2008) claim that this has been used when identifying pairwise epistasis by searching for synthetic lethal double mutants (Tong et al. 2001, 2004; Pan et al. 2004, 2006; Davierwala et al. 2005). A fourth is the “Log” definition presented by Mani et al. (2008) and Sanjuan and Elena (2006). The less-frequently used “scaled ɛ” (Segre et al. 2005) measure of epistasis takes the multiplicative definition of independence with a scaling factor.These different definitions of independence are partly due to distinct measurement “scales.” For some traits, a multiplicative definition of independence may be necessary to identify epistasis between two genes, whereas for other traits, additivity may be appropriate (Falconer and Mackay 1995; Wade et al. 2001; Mani et al. 2008; Phillips 2008). An interaction found under one independence definition may not necessarily be found under another, leading to different biological conclusions (Mani et al. 2008).Mani et al. (2008) suggest that there may be an “ideal” definition of independence for all gene pairs for identifying functional relationships. However, it is plausible that different representations of independence for two genes may reflect different biological properties of the relationship (Kupper and Hogan 1978; Rothman et al. 1980). “Two categories of general interest [the additive and multiplicative definitions, respectively] are those in which etiologic factors act interchangeably in the same step in a multistep process, or alternatively act at different steps in the process” (Rothman et al. 1980, p. 468). In some cases, the discovery of epistasis may merely be an artifact of using an incorrect null model (Kupper and Hogan 1978). It may be necessary to represent “independence” differently, resulting in different statistical measures of interactions, for different pairs of genes depending on their functions.Previous studies have suggested that different pairs of loci may have different modes of interaction and have attempted to subclassify genetic interactions into regulatory hierarchies and mutually exclusive “interaction subtypes” to elucidate underlying biological properties (Avery and Wasserman 1992; Drees et al. 2005; St. Onge et al. 2007). We suggest that epistatic relationships can be divided into several subtypes, or forms, corresponding to the aforementioned definitions of independence. As a particular gene pair may deviate from independence according to several criteria, we do not claim that these subtypes are necessarily mutually exclusive. We attempt to select the most likely epistatic subtype that is the best statistical representation of the relationship between two genes. To further subclassify interactions, epistasis among deleterious mutations can take one of two commonly used forms: positive (equivalently alleviating, antagonistic, or buffering) epistasis, where the phenotype of the double mutant is less severe than expected under independence, and negative (equivalently aggravating, synergistic, or synthetic), where the phenotype is more severe than expected (Segre et al. 2005; Collins et al. 2006; Desai et al. 2007; Mani et al. 2008).Another objective of such distinctions is to reduce the bias of the estimator of the epistatic parameter (ɛ), which measures the extent and direction of epistasis for a given gene pair. Mani et al. (2008), assuming that the overall distribution of ɛ should be centered around 0, find that inaccurately choosing a definition of independence can result in increased bias when estimating ɛ. For example, using the minimum definition results in the most severe bias when single mutants have moderate fitness effects, and the additive definition results in the largest positive bias when at least one gene has an extreme fitness defect (Mani et al. 2008). Therefore, it is important to select an optimal estimator for ɛ for each pair of genes from among the subtypes of epistatic interactions.Epistasis may be important to consider in genomic association studies, as a gene with a weak main effect may be identified only through its interaction with another gene or other genes (Frankel and Schork 1996; Culverhouse et al. 2002; Moore 2003; Cordell 2009; Moore and Williams 2009). Epistasis has also been studied extensively in the context of the evolution of sex and recombination. The mutational deterministic hypothesis proposes that the evolution of sex and recombination would be favored by negative epistatic interactions (Feldman et al. 1980; Kondrashov 1994); many other studies have also studied the importance of the form of epistasis (Elena and Lenski 1997; Otto and Feldman 1997; Burch and Chao 2004; Keightley and Otto 2006; Desai et al. 2007; MacCarthy and Bergman 2007). Indeed, according to Mani et al. (2008, p. 3466), “the choice of definition [of epistasis] alters conclusions relevant to the adaptive value of sex and recombination.”Given fitness data from single and double mutants in haploid organisms, we implement a likelihood method to determine the subtype that is the best statistical representation of the epistatic interaction for pairs of genes. We use maximum-likelihood estimation and the Bayesian information criteria (BIC) (Schwarz 1978) with a likelihood-ratio test to select the most appropriate null or epistatic model for each putative interaction. We conduct extensive simulations to gauge the performance of our method and demonstrate that it performs reasonably well under various interaction scenarios. We apply our method to two data sets with fitness measurements obtained from yeast (Jasnos and Korona 2007; St. Onge et al. 2007), whose authors assume only multiplicative epistasis for all interactions. By examining functional links and experimentally validated interactions among epistatic pairs, we demonstrate that our results are biologically meaningful. Studying a random selection of genes, we find that minimum epistasis is more prevalent than both additive and multiplicative epistasis and that the overall distribution of ɛ is not significantly different from zero (as Jasnos and Korona 2007 suggest). For genes in a particular pathway, we advise selecting among fewer epistatic subtypes. We believe that our method of epistatic subtype classification will aid in understanding genetic interactions and their properties.

St. Onge et al. (2007) data set:

St. Onge et al. (2007) examined 26 nonessential genes known to confer resistance to MMS, constructed double-deletion strains for 323 double-mutant strains (all but two of the total possible pairs), and assumed the multiplicative form of epistasis for all interactions (see Methods: Analysis of experimental data). Following these authors, we focus on single- and double-mutant fitnesses measured in the presence of MMS. (For results in the absence of MMS, see File S1 and File S1_2.)Using the resampling method described in Analysis of experimental data and File S1, 222 gene pairs pass the cutoff of having epistasis inferred in at least 900 of 1000 replicates. This does not include 5 synthetic lethal gene pairs. Hypothesis testing and a multiple-testing procedure (for 222 simultaneous hypotheses) are necessary to determine the final epistatic pairs.To select one among the three multiple-testing procedures, we follow St. Onge et al. (2007) and examine gene pairs that share specific functional links (see Analysis of experimental data). The Bonferroni method is likely too conservative, yielding only 25 significantly epistatic pairs with only one functional link among them; alternatively, the pFDR procedure appears to be too lenient in rejecting independence for all 222 pairs. Therefore, we use the FDR procedure (although the number of functional links is not significant) and detect 193 epistatic pairs, of which 5 (2.6%) are synthetic lethals, 19 (9.8%) have additive epistasis, 33 (17.1%) have multiplicative epistasis, and 136 (70.5%) have minimum epistasis (File S1_1). We find 29 gene pairs with positive (alleviating) epistasis and 159 gene pairs with negative (aggravating) epistasis.

TABLE 2

Summary of gene pairs with the indicated epistatic subtypes, inferred using the FDR procedure with the BIC method that considers all three epistatic subtypes and their corresponding null models
Epistatic subtypeStudy SStudy J
All193 (100%)352 (100%)
= −0.060 = −0.001
= −0.096 = −0.059
Additive19 (9.8%)35 (9.9%)
= 0.115* = 0.193***
= 0.131 = 0.188
Multiplicative33 (17.1%)63 (17.9%)
= 0.048 = 0.017
= −0.166 = −0.115
Minimum136 (70.5%)254 (72.2%)
= −0.111*** = −0.032**
= −0.091 = −0.065
Open in a separate windowNumbers are the counts of each type, and percentages are given of the total number of epistatic pairs. The mean () and median () of the epistatic parameter (ɛ) are given for each subtype, with “*” indicating that the mean of ɛ is significantly different from 0 (*, P-value ≤0.05; **, P-value ≤0.01; ***, P-value ≤0.001). Study S refers to the St. Onge et al. (2007) data set, and study J refers to the Jasnos and Korona (2007) data set. (For study S, five of the epistatic pairs are synthetic lethals and are not shown; as a result, percentages do not sum to 100%.)To further validate the use of our method and the FDR procedure, we assess by Fisher''s exact test the significance of an enrichment of both Biological Process and all GO Slim term links among epistatic pairs, neither of which are significant (Gene Ontology Consortium 2000; www.yeastgenome.org; Stark et al. 2006); Table S4]. Although some of the previously unidentified interactions that we identify could be false positives, many are likely to be new discoveries.

TABLE 3

Comparison of validation measures for each data set for different variations of the FDR and BIC procedures, considering only a subset of epistatic subtypes with their corresponding null models: all epistatic subtypes (A, P, and M); only the additive and multiplicative subtypes (A and P); and only the additive (A), only the multiplicative (P), or only the minimum (M) subtype (see text for details)
Subtypes considered in BIC procedure
A, P, MA, PAPM
Study J
No. found (636)352273263231329
Functional links (25)19 (0.0255)*13 (0.2320)11 (0.4689)10 (0.4227)15 (0.2619)
GO Slim terms (Biological Process) (115)69 (0.1573)50 (0.4874)55 (0.0736)44 (0.3534)68 (0.04902)*
GO Slim terms (all) (369)224 (0.0009)*172 (0.01654)*160 (0.1297)146 (0.0273)*213 (0.0003)*
Experimentally identified (3)32123
Study S
No. found (323)193192247171243
Functional links (36)21 (0.6450)29 (0.0041)*34 (0.0031)*29 (0.0003)*24 (0.9256)
GO Slim terms (Biological Process) (283)174 (0.0657)174 (0.03656)*223 (0.0010)*153 (0.1825)213 (0.5534)
GO Slim terms (all) (307)185 (0.2866)182 (0.6926)237 (0.1472)162 (0.6997)231 (0.5908)
Experimentally identified (29)1722242321
Open in a separate windowNumbers in parentheses indicate P-values by Fisher''s exact test. “*” indicates significance. Study J refers to the Jasnos and Korona (2007) data set, and study S refers to the St. Onge et al. (2007) data set measured in the presence of MMS. Numbers in parentheses indicate the total number of tested pairs and the total number of each type of link found in each complete data set.The epistatic subtypes we consider are not necessarily mutually exclusive. To more fully assess the assumptions of our method, we also consider several of the possible subsets of the epistatic subtypes (and their corresponding null models) in our procedure. As the minimum epistatic subtype was the most frequently selected in this data set, we first do not include the minimum null model or the minimum epistatic model in our procedure (i.e., we select from among four rather than six models for a pair; Table S4). However, there are a significant number of epistatic pairs with functional links only when the minimum epistatic subtype is not included (also see Table S4 and Table S5). It is not immediately clear which epistatic subtypes are the most appropriate for these data, although including the minimum subtype may not be appropriate (Mani et al. 2008) (see discussion).Although it may be best to consider fewer epistatic subtypes for this specific data set, we report our results including all three epistatic subtypes and their corresponding null models (St. Onge et al. (2007), although we identify 105 epistatic pairs not identified by the original authors (Figure S4, Table S4). St. Onge et al. (2007) find that epistatic pairs with a functional link have a positively shifted distribution of epistasis. We find no such shift in epistasis values (Figure S5). We also demonstrate [described in application to simulated data: Bias and variance of the epistatic parameter (ɛ)] that our method seems to reduce bias of the epistatic parameter (ɛ) (Table S3).] When considering only a subset of the epistatic subtypes, however, we find to be positive and significantly different from zero (results not shown). See File S1, Figure S6, and Figure S7 for additional discussion of the epistatic pairs we identify.

Jasnos and Korona (2007) data set:

The Jasnos and Korona (2007) data set included 758 yeast gene deletions known to cause growth defects and reports fitnesses of only a sparse subset of all possible gene pairs [≈0.2% of the possible pairwise genotypes, or 639 pairs of ]. Because the authors do not identify epistatic pairs in a hypothesis-testing framework, we cannot explicitly compare our conclusions with theirs.To validate our method, we examine gene pairs that have specific functional links (see methods: Analysis of experimental data). When defining a functional link using GO terms (Gene Ontology Consortium 2000) with <30 genes associated with them, only 1 of 639 tested gene pairs has a functional link. Raising the threshold of associated genes to 50 and 100, the number of tested pairs with functional links rises only to 3 and 9, respectively. Because of the large number of random genes and the sparse number of gene pairs in this data set, we follow Tong et al. (2004) and select GO terms that have associated with them ≤200 genes. Twenty-five of 639 tested pairs then have a functional link.Only the FDR multiple-testing procedure results in a significant enrichment of functional links among epistatic pairs (File S1). With the FDR procedure we find 352 significant epistatic pairs, of which 35 (9.9%) have additive epistasis, 63 (17.9%) have multiplicative epistasis, and 254 (72.2%) have minimum epistasis (File S1_3). These proportions of inferred subtypes suggest that the authors'' original restriction to multiplicative epistasis may be inappropriate. We find 141 gene pairs with positive epistasis and 211 gene pairs with negative epistasis.We do not find a significant number of epistatic pairs with shared GO Slim Biological Process terms (see Analysis of experimental data), but do when considering all shared GO Slim terms (St. Onge et al. (2007) data set, we also consider some of the possible subsets of the three epistatic subtypes (and their corresponding null models) in our model selection procedure (Table S5). In contrast to the St. Onge et al. (2007) data set, using all three epistatic subtypes results in a significant number of epistatic pairs with functional links; this measure is not significant when using any of the other subsets of the subtypes. This suggests that our proposed method with three epistatic subtypes may indeed be the most appropriate for data sets with randomly selected genes.We examined the distribution of the estimated values of the epistatic parameter (ɛ) for all pairs with significant epistasis. Jasnos and Korona (2007), in assuming only multiplicative epistasis, conclude that epistasis is predominantly positive. However, we find that the estimated mean of epistasis is not significantly different from zero (two-sided t-test, P-value = 0.9578; Figure 1 and File S1.Open in a separate windowFigure 1.—Distribution of the epistasis values (ɛ) for significant epistatic pairs in the Jasnos and Korona (2007) data set, determined using the FDR procedure and the BIC method including all three epistatic subtypes and their corresponding null models. Mean of ɛ is −0.0009, with a standard deviation of 0.3177; median value is −0.0587. A similar plot is shown in Figure 3 of Jasnos and Korona (2007).  相似文献   

8.
The capacity for phenotypic evolution is dependent upon complex webs of functional interactions that connect genotype and phenotype. Wrinkly spreader (WS) genotypes arise repeatedly during the course of a model Pseudomonas adaptive radiation. Previous work showed that the evolution of WS variation was explained in part by spontaneous mutations in wspF, a component of the Wsp-signaling module, but also drew attention to the existence of unknown mutational causes. Here, we identify two new mutational pathways (Aws and Mws) that allow realization of the WS phenotype: in common with the Wsp module these pathways contain a di-guanylate cyclase-encoding gene subject to negative regulation. Together, mutations in the Wsp, Aws, and Mws regulatory modules account for the spectrum of WS phenotype-generating mutations found among a collection of 26 spontaneously arising WS genotypes obtained from independent adaptive radiations. Despite a large number of potential mutational pathways, the repeated discovery of mutations in a small number of loci (parallel evolution) prompted the construction of an ancestral genotype devoid of known (Wsp, Aws, and Mws) regulatory modules to see whether the types derived from this genotype could converge upon the WS phenotype via a novel route. Such types—with equivalent fitness effects—did emerge, although they took significantly longer to do so. Together our data provide an explanation for why WS evolution follows a limited number of mutational pathways and show how genetic architecture can bias the molecular variation presented to selection.UNDERSTANDING—and importantly, predicting—phenotypic evolution requires knowledge of the factors that affect the translation of mutation into phenotypic variation—the raw material of adaptive evolution. While much is known about mutation rate (e.g., Drake et al. 1998; Hudson et al. 2002), knowledge of the processes affecting the translation of DNA sequence variation into phenotypic variation is minimal.Advances in knowledge on at least two fronts suggest that progress in understanding the rules governing the generation of phenotypic variation is possible (Stern and Orgogozo 2009). The first stems from increased awareness of the genetic architecture underlying specific adaptive phenotypes and recognition of the fact that the capacity for evolutionary change is likely to be constrained by this architecture (Schlichting and Murren 2004; Hansen 2006). The second is the growing number of reports of parallel evolution (e.g., Pigeon et al. 1997; ffrench-Constant et al. 1998; Allender et al. 2003; Colosimo et al. 2004; Zhong et al. 2004; Boughman et al. 2005; Shindo et al. 2005; Kronforst et al. 2006; Woods et al. 2006; Zhang 2006; Bantinaki et al. 2007; McGregor et al. 2007; Ostrowski et al. 2008)—that is, the independent evolution of similar or identical features in two or more lineages—which suggests the possibility that evolution may follow a limited number of pathways (Schluter 1996). Indeed, giving substance to this idea are studies that show that mutations underlying parallel phenotypic evolution are nonrandomly distributed and typically clustered in homologous genes (Stern and Orgogozo 2008).While the nonrandom distribution of mutations during parallel genetic evolution may reflect constraints due to genetic architecture, some have argued that the primary cause is strong selection (e.g., Wichman et al. 1999; Woods et al. 2006). A means of disentangling the roles of population processes (selection) from genetic architecture is necessary for progress (Maynard Smith et al. 1985; Brakefield 2006); also necessary is insight into precisely how genetic architecture might bias the production of mutations presented to selection.Despite their relative simplicity, microbial populations offer opportunities to advance knowledge. The wrinkly spreader (WS) morphotype is one of many different niche specialist genotypes that emerge when experimental populations of Pseudomonas fluorescens are propagated in spatially structured microcosms (Rainey and Travisano 1998). Previous studies defined, via gene inactivation, the essential phenotypic and genetic traits that define a single WS genotype known as LSWS (Spiers et al. 2002, 2003) (Figure 1). LSWS differs from the ancestral SM genotype by a single nonsynonymous nucleotide change in wspF. Functionally (see Figure 2), WspF is a methyl esterase and negative regulator of the WspR di-guanylate cyclase (DGC) (Goymer et al. 2006) that is responsible for the biosynthesis of c-di-GMP (Malone et al. 2007), the allosteric activator of cellulose synthesis enzymes (Ross et al. 1987). The net effect of the wspF mutation is to promote physiological changes that lead to the formation of a microbial mat at the air–liquid interface of static broth microcosms (Rainey and Rainey 2003).Open in a separate windowFigure 1.—Outline of experimental strategy for elucidation of WS-generating mutations and their subsequent identity and distribution among a collection of independently evolved, spontaneously arising WS genotypes. The strategy involves, first, the genetic analysis of a specific WS genotype (e.g., LSWS) to identify the causal mutation, and second, a survey of DNA sequence variation at specific loci known to harbor causal mutations among a collection of spontaneously arising WS genotypes. For example, suppressor analysis of LSWS using a transposon to inactivate genes necessary for expression of the wrinkly morphology delivered a large number of candidate genes (top left) (Spiers et al. 2002). Genetic and functional analysis of these candidate genes (e.g., Goymer et al. 2006) led eventually to the identity of the spontaneous mutation (in wspF) responsible for the evolution of LSWS from the ancestral SM genotype (Bantinaki et al. 2007). Subsequent analysis of the wspF sequence among 26 independent WS genotypes (bottom) showed that 50% harbored spontaneous mutations (of different kinds; see Open in a separate windowFigure 2.—Network diagram of DGC-encoding pathways underpinning the evolution of the WS phenotype and their regulation. Overproduction of c-di-GMP results in overproduction of cellulose and other adhesive factors that determine the WS phenotype. The ancestral SBW25 genome contains 39 putative DGCs, each in principle capable of synthesizing the production of c-di-GMP, and yet WS genotypes arise most commonly as a consequence of mutations in just three DGC-containing pathways: Wsp, Aws, and Mws. In each instance, the causal mutations are most commonly in the negative regulatory component: wspF, awsX, and the phosphodiesterase domain of mwsR (see text).To determine whether spontaneous mutations in wspF are a common cause of the WS phenotype, the nucleotide sequence of this gene was obtained from a collection of 26 spontaneously arising WS genotypes (WSA-Z) taken from 26 independent adaptive radiations, each founded by the same ancestral SM genotype (Figure 1): 13 contained mutations in wspF (Bantinaki et al. 2007). The existence of additional mutational pathways to WS provided the initial motivation for this study.

TABLE 1

Mutational causes of WS
WS genotypeGeneNucleotide changeAmino acid changeSource/reference
LSWSwspFA901CS301RBantinaki et al. (2007)
AWSawsXΔ100-138ΔPDPADLADQRAQAThis study
MWSmwsRG3247AE1083KThis study
WSAwspFT14GI5SBantinaki et al. (2007)
WSBwspFΔ620-674P206Δ (8)aBantinaki et al. (2007)
WSCwspFG823TG275CBantinaki et al. (2007)
WSDwspEA1916GD638GThis study
WSEwspFG658TV220LBantinaki et al. (2007)
WSFwspFC821TT274IBantinaki et al. (2007)
WSGwspFC556TH186YBantinaki et al. (2007)
WSHwspEA2202CK734NThis study
WSIwspEG1915TD638YThis study
WSJwspFΔ865-868R288Δ (3)aBantinaki et al. (2007)
WSKawsOG125TG41VThis study
WSLwspFG482AG161DBantinaki et al. (2007)
WSMawsRC164TS54FThis study
WSNwspFA901CS301RBantinaki et al. (2007)
WSOwspFΔ235-249V79Δ (6)aBantinaki et al. (2007)
WSPawsR222insGCCACCGAA74insATEThis study
WSQmwsR3270insGACGTG1089insDVThis study
WSRmwsRT2183CV272AThis study
WSSawsXC472TQ158STOPThis study
WSTawsXΔ229-261ΔYTDDLIKGTTQThis study
WSUwspFΔ823-824T274Δ (13)aBantinaki et al. (2007)
WSVawsXT74GL24RThis study
WSWwspFΔ149L49Δ (1)aBantinaki et al. (2007)
WSXb???This study
WSYwspFΔ166-180Δ(L51-I55)Bantinaki et al. (2007)
WSZ
mwsR
G3055A
A1018T
This study
Open in a separate windowaP206Δ(8) indicates a frameshift; the number of new residues before a stop codon is reached is in parentheses.bSuppressor analysis implicates the wsp locus (17 transposon insertions were found in this locus). However, repeated sequencing failed to identify a mutation.Here we define and characterize two new mutational routes (Aws and Mws) that together with the Wsp pathway account for the evolution of 26 spontaneously arising WS genotypes. Each pathway offers approximately equal opportunity for WS evolution; nonetheless, additional, less readily realized genetic routes producing WS genotypes with equivalent fitness effects exist. Together our data show that regulatory pathways with specific functionalities and interactions bias the molecular variation presented to selection.  相似文献   

9.
Rönnegård L  Valdar W 《Genetics》2011,188(2):435-447
Traditional methods for detecting genes that affect complex diseases in humans or animal models, milk production in livestock, or other traits of interest, have asked whether variation in genotype produces a change in that trait’s average value. But focusing on differences in the mean ignores differences in variability about that mean. The robustness, or uniformity, of an individual’s character is not only of great practical importance in medical genetics and food production but is also of scientific and evolutionary interest (e.g., blood pressure in animal models of heart disease, litter size in pigs, flowering time in plants). We describe a method for detecting major genes controlling the phenotypic variance, referring to these as vQTL. Our method uses a double generalized linear model with linear predictors based on probabilities of line origin. We evaluate our method on simulated F2 and collaborative cross data, and on a real F2 intercross, demonstrating its accuracy and robustness to the presence of ordinary mean-controlling QTL. We also illustrate the connection between vQTL and QTL involved in epistasis, explaining how these concepts overlap. Our method can be applied to a wide range of commonly used experimental crosses and may be extended to genetic association more generally.QUANTITATIVE trait locus (QTL) analysis has traditionally focused on detection of major genes controlling the expected mean of a phenotype. But there is substantial evidence that not only the mean but also the variance, that is, the stochastic variability of the phenotype about its average value, may itself be under genetic control. The identification of such variance-controlling loci, which we call vQTL, can be helpful in a variety of contexts, including selection of livestock for uniformity, evaluating predictability of response to medical treatment, identification of key biomolecular stabilizers, and assessment of population resilience in ecology and evolution.One way of interpreting an increase in variability is as a decrease in stability. Waddington (1942) described the concept of canalization, whereby natural selection favors the relative constancy of some attributes, for example, well-formed organs and limbs, and thereby leads to the evolution of heritable architectures that buffer the impact of environmental or background genetic variation that would otherwise cause development to go astray. These architectures create virtual “canals” down which developmental programs flow. For a canalized phenotype, which modern usage expands to include nondevelopmental traits, the “zone of canalization” is the range of underlying liability over which potentially disruptive variation may be absorbed without serious consequence to the expressed trait value (Lynch and Walsh 1998). A well-studied example of a stabilizing architecture is that provided by heat-shock protein 90 (Hsp90), which buffers genetic and stochastic variation in the development of plants and flies (Rutherford and Lindquist 1998; Queitsch et al. 2002; Sangster et al. 2008).But in absorbing variation, such stabilizing architectures also hide it from view, and a sensitizing change in the stabilizer that shifts liability outside the zone of canalization can have a dramatic effect on the phenotype. Such shifts release the combined effects of previously “cryptic” genetic variation: now decanalized, the phenotype is more sensitive to internal (including genetic) and external environment, and as a result varies more greatly between individuals (Dworkin 2005; Hornstein and Shomron 2006). In this vein, decanalization has been proposed to explain why the genetic architectures of some diseases in human populations seem more amenable than others to genetic dissection through genome-wide association (Gibson and Goldstein 2007). Specifically, whereas some disease phenotypes in homogeneous populations may be heavily canalized and thereby harder to dissect, others may have been decanalized by modern living conditions (e.g., inflammatory diseases) or modern admixture, while yet others are simply too recent in evolutionary history for buffering networks to have evolved (e.g., response to HIV).Increased variability can also be adaptive. In natural populations disruptive selection favors diversity, with increased “capacitance” (Rice 2008) or “bet-hedging” (Beaumont et al. 2009) spreading risk over a variable fitness landscape. Feinberg and Irizarry (2010) recently proposed a heritable and selectable mechanism for this based on stochastic epigenetic variation. In controlled populations, variability can be increased through directional selection. For example, in a Drosophila selection experiment Clayton and Robertson (1957) reported increased bristle number variance, which is consistent with the idea that genotypes associated with higher environmental variance have a greater chance of being selected under directional selection (Hill and Zhang 2004). Moreover, genetic differences have been observed for phenotypic variability in body weight for chickens (Rowe et al. 2006) and snails (Ros et al. 2004) and litter size in rabbits (Ibanez-Escriche et al. 2008), sheep (Sancristobal-Gaudy et al. 1998), and pigs (Sorensen and Waagepetersen 2003).In natural populations with stabilizing selection we should expect to find alleles minimizing variance for fitness traits (Lande 1980; Houle 1992), whereas directional selection during domestication will favor alleles that increase variance. One may therefore expect to find vQTL in experimental crosses between wild and domestic animals (see Andersson 2001). Nonetheless, genetic buffering that leads to phenotypic robustness need not require an evolutionary explanation to be observed, nor to be useful in medicine and agriculture. Plainly, detecting vQTL and inferring how they arose are separate questions; here we concentrate on the first.
Sources of phenotypic variability
Variance groupaDecanalization (epistasis)Environmental sensitivityTemporal fluctuationMeasurement error
Genetically distinct individuals with same allele at a vQTLb
Genetically identical individuals
Same individual at different times
Same individual at the same time
Open in a separate windowaThe group in which variance is assessed, and between which variance is compared.bThe variance groups compared here.Few studies have explicitly looked for vQTL. Among the more recent, Ordas et al. (2008) studied morphological traits and flowering time in maize. They detected vQTL by contrasting the residual variance between genotypes in replicates of recombinant inbred lines (RILs; see second row, Wittenburg et al. (2009) examined the sample variance of birth weight within pig litters as a gamma-distributed trait among 3914 sows, estimating a heritability of 0.1 for this trait using a generalized linear mixed model. Sangster et al. (2008) used Levene''s test for detection of variance-controlling genes. In that test, the absolute values of the residuals are used as a response in an ANOVA (e.g., Faraway 2004). Mackay and Lyman (2005) studied Drosophila bristle number and found substantial differences in the coefficient of variation (CV) between inbred lines, comparing CV also using ANOVA. The methods used in these last two studies have the limitation of not being able to model confounding effects in the mean. Using residuals (as in Sangster et al. 2008; Wittenburg et al. 2009) can potentially incorporate covariates but involves conditioning on unknowns. There is thus considerable utility in a method that simultaneously estimates means and variances, flexibly accommodates covariates, applies to a wide range of experimental crosses, and is robust and fast enough for genome-wide analyses.Regression-based models (Haley and Knott 1992; Martinez and Curnow 1992) have proven to be fast and powerful at detecting QTL controlling the mean of a complex trait in experimental crosses and flexible since they are straightforwardly extended to include epistatic effects and interactions (Carlborg and Haley 2004). Mott et al. (2000) developed the haplotype reconstruction method HAPPY and its associated regression model, which allows for a variable number of strains and may therefore be applied to vQTL mapping in, e.g., heterogeneous stocks (HS; Valdar et al. 2006,b) and multiparent advanced generation inbred cross resource populations (MAGIC lines; Cavanagh et al. 2008) such as the collaborative cross (CC; Churchill et al. 2004; Broman 2005; Valdar et al. 2006a; Chesler et al. 2008) and the Arabidopsis recombinant inbred lines of Kover et al. (2009).Our aim is to develop a regression model for detection of major genes controlling phenotypic variance that can be applied genome wide. The estimation uses double generalized linear models (DGLMs; Smyth 1989) and its parameterization is based on the HAPPY formulation of inferred haplotypes. The method fits ordinary QTL and vQTL simultaneously in the same model. We apply it to simulated data from an F2 and the CC and real data from an F2 intercross of partially inbred lines.  相似文献   

10.
The Conserved miR-51 microRNA Family Is Redundantly Required for Embryonic Development and Pharynx Attachment in Caenorhabditis elegans     
W. Robert Shaw  Javier Armisen  Nicolas J. Lehrbach  Eric A. Miska 《Genetics》2010,185(3):897-905
A major question about cytokinesis concerns the role of the septin proteins, which localize to the division site in all animal and fungal cells but are essential for cytokinesis only in some cell types. For example, in Schizosaccharomyces pombe, four septins localize to the division site, but deletion of the four genes produces only a modest delay in cell separation. To ask if the S. pombe septins function redundantly in cytokinesis, we conducted a synthetic-lethal screen in a septin-deficient strain and identified seven mutations. One mutation affects Cdc4, a myosin light chain that is an essential component of the cytokinetic actomyosin ring. Five others cause frequent cell lysis during cell separation and map to two loci. These mutations and their dosage suppressors define a signaling pathway (including Rho1 and a novel arrestin) for repairing cell-wall damage. The seventh mutation affects the poorly understood RNA-binding protein Scw1 and severely delays cell separation when combined either with a septin mutation or with a mutation affecting the septin-interacting, anillin-like protein Mid2, suggesting that Scw1 functions in a pathway parallel to that of the septins. Taken together, our results suggest that the S. pombe septins participate redundantly in one or more pathways that cooperate with the actomyosin ring during cytokinesis and that a septin defect causes septum defects that can be repaired effectively only when the cell-integrity pathway is intact.THE fission yeast Schizosaccharomyces pombe provides an outstanding model system for studies of cytokinesis (McCollum and Gould 2001; Balasubramanian et al. 2004; Pollard and Wu 2010). As in most animal cells, successful cytokinesis in S. pombe requires an actomyosin ring (AMR). The AMR begins to assemble at the G2/M transition and involves the type II myosin heavy chains Myo2 and Myp2 and the light chains Cdc4 and Rlc1 (Wu et al. 2003). Myo2 and Cdc4 are essential for cytokinesis under all known conditions, Rlc1 is important at all temperatures but essential only at low temperatures, and Myp2 is essential only under stress conditions. As the AMR constricts, a septum of cell wall is formed between the daughter cells. The primary septum is sandwiched by secondary septa and subsequently digested to allow cell separation (Humbel et al. 2001; Sipiczki 2007). Because of the internal turgor pressure of the cells, the proper assembly and structural integrity of the septal layers are essential for cell survival.Septum formation involves the β-glucan synthases Bgs1/Cps1/Drc1, Bgs3, and Bgs4 (Ishiguro et al. 1997; Le Goff et al. 1999; Liu et al. 1999, 2002; Martín et al. 2003; Cortés et al. 2005) and the α-glucan synthase Ags1/Mok1 (Hochstenbach et al. 1998; Katayama et al. 1999). These synthases are regulated by the Rho GTPases Rho1 and Rho2 and the protein kinase C isoforms Pck1 and Pck2 (Arellano et al. 1996, 1997, 1999; Nakano et al. 1997; Hirata et al. 1998; Calonge et al. 2000; Sayers et al. 2000; Ma et al. 2006; Barba et al. 2008; García et al. 2009b). The Rho GTPases themselves appear to be regulated by both GTPase-activating proteins (GAPs) and guanine-nucleotide-exchange factors (GEFs) (Nakano et al. 2001; Calonge et al. 2003; Iwaki et al. 2003; Tajadura et al. 2004; Morrell-Falvey et al. 2005; Mutoh et al. 2005; García et al. 2006, 2009a,b). In addition, septum formation and AMR function appear to be interdependent. In the absence of a normal AMR, cells form aberrant septa and/or deposit septal materials at random locations, whereas a mutant defective in septum formation (bgs1) is also defective in AMR constriction (Gould and Simanis 1997; Le Goff et al. 1999; Liu et al. 1999, 2000). Both AMR constriction and septum formation also depend on the septation initiation network involving the small GTPase Spg1 (McCollum and Gould 2001; Krapp and Simanis 2008). Despite this considerable progress, many questions remain about the mechanisms and regulation of septum formation and its relationships to the function of the AMR.One major question concerns the role(s) of the septins. Proteins of this family are ubiquitous in fungal and animal cells and typically localize to the cell cortex, where they appear to serve as scaffolds and diffusion barriers for other proteins that participate in a wide variety of cellular processes (Longtine et al. 1996; Gladfelter et al. 2001; Hall et al. 2008; Caudron and Barral 2009). Despite the recent progress in elucidating the mechanisms of septin assembly (John et al. 2007; Sirajuddin et al. 2007; Bertin et al. 2008; McMurray and Thorner 2008), the details of septin function remain obscure. However, one prominent role of the septins and associated proteins is in cytokinesis. Septins concentrate at the division site in every cell type that has been examined, and in Saccharomyces cerevisiae (Hartwell 1971; Longtine et al. 1996; Lippincott et al. 2001; Dobbelaere and Barral 2004) and at least some Drosophila (Neufeld and Rubin 1994; Adam et al. 2000) and mammalian (Kinoshita et al. 1997; Surka et al. 2002) cell types, the septins are essential for cytokinesis. In S. cerevisiae, the septins are required for formation of the AMR (Bi et al. 1998; Lippincott and Li 1998). However, this cannot be their only role, because the AMR itself is not essential for cytokinesis in this organism (Bi et al. 1998; Korinek et al. 2000; Schmidt et al. 2002). Moreover, there is no evidence that the septins are necessary for AMR formation or function in any other organism. A further complication is that in some cell types, including most Caenorhabditis elegans cells (Nguyen et al. 2000; Maddox et al. 2007) and some Drosophila cells (Adam et al. 2000; Field et al. 2008), the septins do not appear to be essential for cytokinesis even though they localize to the division site.S. pombe has seven septins, four of which (Spn1, Spn2, Spn3, and Spn4) are expressed in vegetative cells and localize to the division site shortly before AMR constriction and septum formation (Longtine et al. 1996; Berlin et al. 2003; Tasto et al. 2003; Wu et al. 2003; An et al. 2004; Petit et al. 2005; Pan et al. 2007; Onishi et al. 2010). Spn1 and Spn4 appear to be the core members of the septin complex (An et al. 2004; McMurray and Thorner 2008), and mutants lacking either of these proteins do not assemble the others at the division site. Assembly of a normal septin ring also depends on the anillin-like protein Mid2, which colocalizes with the septins (Berlin et al. 2003; Tasto et al. 2003). Surprisingly, mutants lacking the septins are viable and form seemingly complete septa with approximately normal timing. These mutants do, however, display a variable delay in separation of the daughter cells, suggesting that the septins play some role(s) in the proper completion of the septum or in subsequent processes necessary for cell separation (Longtine et al. 1996; An et al. 2004; Martín-Cuadrado et al. 2005).It is possible that the septins localize to the division site and yet are nonessential for division in some cell types because their role is redundant with that of some other protein(s) or pathway(s). To explore this possibility in S. pombe, we screened for mutations that were lethal in combination with a lack of septins. The results suggest that the septins cooperate with the AMR during cytokinesis and that, in the absence of septin function, the septum is not formed properly, so that an intact system for recognizing and repairing cell-wall damage becomes critical for cell survival.  相似文献   

11.
Stb3 Plays a Role in the Glucose-Induced Transition from Quiescence to Growth in Saccharomyces cerevisiae     
Dritan Liko  Michael K. Conway  Douglas S. Grunwald  Warren Heideman 《Genetics》2010,185(3):797-810
  相似文献   

12.
Snf1p Regulates Gcn5p Transcriptional Activity by Antagonizing Spt3p     
Yang Liu  Xinjing Xu  Min-Hao Kuo 《Genetics》2010,184(1):91-105
  相似文献   

13.
The Genealogical Consequences of Fecundity Variance Polymorphism          下载免费PDF全文
Jesse E. Taylor 《Genetics》2009,182(3):813-837
The genealogical consequences of within-generation fecundity variance polymorphism are studied using coalescent processes structured by genetic backgrounds. I show that these processes have three distinctive features. The first is that the coalescent rates within backgrounds are not jointly proportional to the infinitesimal variance, but instead depend only on the frequencies and traits of genotypes containing each allele. Second, the coalescent processes at unlinked loci are correlated with the genealogy at the selected locus; i.e., fecundity variance polymorphism has a genomewide impact on genealogies. Third, in diploid models, there are infinitely many combinations of fecundity distributions that have the same diffusion approximation but distinct coalescent processes; i.e., in this class of models, ancestral processes and allele frequency dynamics are not in one-to-one correspondence. Similar properties are expected to hold in models that allow for heritable variation in other traits that affect the coalescent effective population size, such as sex ratio or fecundity and survival schedules.THE population genetics of within-generation fecundity variance has been studied from two perspectives. Beginning with Wright (1938), several authors have investigated the relationship between the effective size of a panmictic population with seasonal reproduction and the variance of the number of offspring born to each adult within a season (Crow and Denniston 1988; Nunney 1993, 1996; Waples 2002; Hedrick 2005; Engen et al. 2007). Although the precise form of this relationship depends on other biological factors such as the mating system and the manner in which population regulation operates, each of these studies shows that the effective population size is a decreasing function of fecundity variance. Furthermore, provided that the variance and the coalescent effective population sizes coincide (Ewens 1982; Nordborg and Krone 2002; Sjodin et al. 2005), these results imply that both the rate at which neutral allele frequencies fluctuate from generation to generation and the rate at which lineages coalesce will be positively correlated with within-generation fecundity variance. For example, it has been suggested that the shallow genealogies that have been documented in many marine organisms are a consequence of the high variance of reproductive success in the recruitment sweepstakes operating in these species (Hedgecock 1994; Árnason 2004; Eldon and Wakeley 2006).These results hold in models in which all individuals have the same within-generation (or within-season) fecundity variance. However, the evolutionary genetics of populations that are polymorphic for alleles that influence demographic traits have also been investigated. The first results of this kind were derived by Gillespie (1974, 1975, 1977), who used diffusion theory to show that natural selection can act directly on within-generation fecundity variance in a haploid population with nonoverlapping generations. By studying a simple model of a population composed of two genotypes, say A1 and A2, Gillespie (1974) showed that the fluctuations in the frequency of allele A1 can be approximated by a diffusion process with the following drift and variance coefficients,where p is the frequency of A1, N is the number of adults, and 1 + μi and are the mean and the variance, respectively, of the number of offspring produced by an individual of type Ai. Most discussions of this class of models have focused on the fitness consequences of differences in fecundity variance, which are summarized by the drift coefficient, m(p), of the diffusion approximation. There are two main conclusions. The first is that because m(p) is an increasing function of the difference − , selection can favor alleles that reduce within-generation fecundity variance even if these have lower mean fecundity. Such variance–mean trade-offs can be interpreted as a kind of bet hedging and could explain the evolution of certain risk-spreading traits such as insect oviposition onto multiple host plants (Root and Kareiva 1986) or multiple mating by females (Sarhan and Kokko 2007). On the other hand, because the strength of selection on fecundity variance is inversely proportional to population size, selection for mean–variance trade-offs will usually be dominated by changes in mean fecundity. For this reason, it has been suggested that within-generation bet hedging will be favored only in very small populations (Seger and Brockman 1987; Hopper et al. 2003), although recent theoretical studies have shown that bet hedging can evolve under less restrictive conditions in subdivided populations (Shpak 2005; Lehmann and Balloux 2007; Shpak and Proulx 2007).Less consideration has been given to the diffusion coefficient, v(p), which differs from the familiar quadratic term, p(1 − p), of the Wright–Fisher diffusion. Because the variance effective population size of a monomorphic population depends on the fecundity variance, it is not surprising that v(p) has an additional dependence on the frequency of A1 whenever the two alleles have different offspring variances. However, as noted by Gillespie (1974), the relationship between allele frequency fluctuations and the allelic composition of the population is counterintuitive. For example, when p is close to 1, so that the population is composed mainly of A1-type individuals, the rate of allele frequency fluctuations is dominated by the variance of the A2 genotype. In particular, if we define the variance effective population size by the expression Np(1 − p)/v(p) (Ewens 1982), then not only is this quantity frequency dependent, but also it depends on the life history traits of the missing genotype whenever the population is fixed for one of the two alleles. In contrast, the coalescent effective population size of a monomorphic population depends only on the offspring distribution of the fixed allele. The discrepancy between these two quantities raises the following question: namely, How does fecundity variance polymorphism affect the statistical properties of the genealogy of a random sample of individuals?The answer to this question is of interest for several reasons. First, although the effects of selection on genealogies have received considerable attention (Przeworski et al. 1999; Williamson and Orive 2002; Barton and Etheridge 2004), little is known about the genealogical consequences of variation in traits that alter the coalescent rate. Extrapolating from models in which the effective population size varies under the control of external factors, we might expect the coalescent process in a model with fecundity variance polymorphism to be a stochastic time change of Kingman''s coalescent. However, the results derived in the next section show that this intuition is usually wrong. The second motivation is more practical. Even if changes in fecundity variance are usually controlled by selection on other traits, the existence of interspecific differences in fecundity variance suggests that there must be periods when populations are polymorphic for alleles that alter the fecundity variance. In these instances, it might be possible to use sequence data to identify the loci responsible for these changes, but to do so will require the development of methods that exploit patterns that are unique to models in which the effective population size depends on the genetic composition of the population. For example, whereas the effects of genetic hitchhiking are usually restricted to linked sites (Maynard Smith and Haigh 1974; Kim and Stephan 2002; Przeworski 2002; Przeworski et al. 2005), we will see later that selective sweeps by mutations that affect fecundity variance would have a genomewide impact on polymorphism.Kingman (1982a,b) showed that the genealogy of a sample of individuals from a panmictic, neutrally evolving population of constant size can be described by a simple stochastic process known as the coalescent (or Kingman''s coalescent). One of the most important properties of Kingman''s coalescent is that it is a Markov process, a fact that is heavily exploited in mathematical analyses and that also allows for efficient simulations of genealogies. Unfortunately, this property generally does not hold in populations composed of nonexchangeable individuals. For example, if there are selective differences between individuals, then although the genealogy of a sample of individuals can still be regarded as a stochastic process, selective interactions between individuals cause this process to also depend on the history of nonancestral lineages. The key to overcoming this difficulty is to embed the genealogical process in a larger process that does satisfy the Markov property. This can be done in two ways. One approach is to embed the coalescent tree within a graphical process called the ancestral selection graph (Krone and Neuhauser 1997; Neuhauser and Krone 1997; Donnelly and Kurtz 1999) in which lineages can either branch, giving rise to pairs of potential ancestors, or coalesce. The intuition behind this construction is that the effects of selection on the genealogy can be accounted for by keeping track of a pool of potential ancestors that includes lineages that have failed to persist due to being outcompeted by individuals of higher fitness. Because the branching rates are linear in the number of lineages, while the coalescence rates are quadratic, this process is certain to reach an ultimate ancestor in finite time. The process can be stopped at this time, and both the ancestral and the genotypic status of individual branches can be resolved by assigning random mutations to the graph and then traversing it from the root to the leaves.The second approach is due to Kaplan et al. (1988), who showed that the genealogical history of a sample of genes under selection can be represented by a structured coalescent process. Here we think of the population as being subdivided into several demes, or genetic backgrounds, consisting of individuals that share the same genotype at the selected locus. Because individuals with the same genotype are exchangeable, the rate of coalescence within a background depends only on the size of the background and the number of ancestral lineages sharing that genotype. In addition, mutations at the selected site will move lineages between backgrounds. To obtain a Markov process, we need to keep track of two kinds of information: (i) the types of the ancestral lineages and (ii) the frequencies of the alleles segregating at the selected locus. Fortunately, because one-dimensional diffusion processes are reversible with respect to their stationary distributions (i.e., the detailed balance conditions are satisfied), the ancestral process of allele frequencies at a locus segregating two alleles has the same law as the forward process. Subsequently, Hudson and Kaplan (1988) showed that the genealogy at a linked neutral locus can be described by a structured coalescent defined in terms of the genetic backgrounds at the selected locus; in this case, recombination between the selected and neutral loci can also move lineages between backgrounds.The objective of this article is to extend the structured coalescent to population genetic models in which within-generation fecundity variance is genotype dependent. (The genealogical consequences of polymorphism affecting between-generation fecundity variance will be described in a separate article.) In these models, exchangeability is violated not only by selective differences between individuals, but also by differences in life history traits that affect coalescent rates and allele frequency fluctuations. Nonetheless, because lineages are exchangeable within backgrounds, the coalescence and substitution rates can still be calculated conditional on the types of the lineages and the genetic composition of the population. In the next two sections, I derive structured coalescent processes that describe the genealogy at a neutral marker locus that is linked to a second locus (the “selected locus”) that affects fecundity variance. This is first done for a haploid model and then extended to a diploid model in which there may be both sex- and genotype-specific differences in fecundity variance. Results for both models are summarized in Rates
TransitionHaploid modelDiploid modeln1μ1q/pn1μ1q/pn2μ2p/qn2μ2p/qn1rqn1rqn2rpn2rp
Open in a separate windowThis work shows that coalescent processes in populations with fecundity variance polymorphism differ from the structured coalescent in a monomorphic population in three ways. One difference is that in populations with fecundity variance polymorphism, the coalescent rates in the different genetic backgrounds are not inversely proportional to the variance effective population size. Instead, coalescence within each allelic background depends only on the frequencies and fecundity distributions of genotypes containing that allele. The second difference is that the genealogies at the marker and selected loci are correlated even when these loci are unlinked; i.e., fecundity variance polymorphism has a genomewide impact on genealogies and genetic variation. This follows from the calculations leading up to Equation 28, which show that the genealogical process at an unlinked marker locus can be represented as a stochastic time change of Kingman''s coalescent dependent on the ancestral process of allele frequencies at the selected locus. The third and most surprising difference is that the correspondence between ancestral processes and allele frequency processes is many-to-one in diploid models with fecundity variance polymorphism. In fact, there are infinitely many combinations of genotype-dependent fecundity distributions (satisfying Equation 24) that have the same diffusion approximation but different genealogical processes. These results are illustrated numerically using simulations of the structured coalescent under directional and balancing selection. Finally, I examine the scope of the theory and some possible applications in the discussion.  相似文献   

14.
Genetic Modifiers of dFMR1 Encode RNA Granule Components in Drosophila          下载免费PDF全文
Anne-Marie J. Cziko  Cathal T. McCann  Iris C. Howlett  Scott A. Barbee  Rebecca P. Duncan  Rene Luedemann  Daniela Zarnescu  Konrad E. Zinsmaier  Roy R. Parker  Mani Ramaswami 《Genetics》2009,182(4):1051-1060
Mechanisms of neuronal mRNA localization and translation are of considerable biological interest. Spatially regulated mRNA translation contributes to cell-fate decisions and axon guidance during development, as well as to long-term synaptic plasticity in adulthood. The Fragile-X Mental Retardation protein (FMRP/dFMR1) is one of the best-studied neuronal translational control molecules and here we describe the identification and early characterization of proteins likely to function in the dFMR1 pathway. Induction of the dFMR1 in sevenless-expressing cells of the Drosophila eye causes a disorganized (rough) eye through a mechanism that requires residues necessary for dFMR1/FMRP''s translational repressor function. Several mutations in dco, orb2, pAbp, rm62, and smD3 genes dominantly suppress the sev-dfmr1 rough-eye phenotype, suggesting that they are required for dFMR1-mediated processes. The encoded proteins localize to dFMR1-containing neuronal mRNPs in neurites of cultured neurons, and/or have an effect on dendritic branching predicted for bona fide neuronal translational repressors. Genetic mosaic analyses indicate that dco, orb2, rm62, smD3, and dfmr1 are dispensable for translational repression of hid, a microRNA target gene, known to be repressed in wing discs by the bantam miRNA. Thus, the encoded proteins may function as miRNA- and/or mRNA-specific translational regulators in vivo.THE subcellular localization and regulated translation of stored mRNAs contributes to cellular asymmetry and subcellular specialization (Lecuyer et al. 2007; Martin and Ephrussi 2009). In mature neurons, local protein synthesis at active synapses may contribute to synapse-specific plasticity that underlies persistent forms of memory (Casadio et al. 1999; Ashraf et al. 2006; Sutton and Schuman 2006; Richter and Klann 2009). During this process, synaptic activity causes local translation of mRNAs normally stored in translationally repressed synaptic mRNPs (Sutton and Schuman 2006; Richter and Klann 2009). While specific neuronal translational repressors and microRNAs have been implicated in this process, their involvement in local translation that underlies memory, as well as the underlying mechanisms, are generally not well understood (Schratt et al. 2006; Keleman et al. 2007; Kwak et al. 2008; Li et al. 2008; Richter and Klann 2009). Furthermore, it remains possible that there are neuron-specific, mRNA-specific, and stimulus-pattern specific pathways for neuronal translational control (Raab-Graham et al. 2006; Giorgi et al. 2007).The Fragile-X Mental Retardation protein (FMRP) is among the best studied of neuronal translational repressors, in part due to its association with human neurodevelopmental disease (Pieretti et al. 1991; Mazroui et al. 2002; Gao 2008). Consistent with function in synaptic translation required for memory formation, mutations in FMRP are associated with increased synaptic translation, enhanced LTD, increased synapse growth, and also with enhanced long-term memory (Zhang et al. 2001; Huber et al. 2002; Bolduc et al. 2008; Dictenberg et al. 2008).FMRP co-immunoprecipitates with components of the RNAi and miRNA machinery and appears to be required for aspects of miRNA function in neurons (Caudy et al. 2002; Ishizuka et al. 2002; Jin et al. 2004b; Gao 2008). In addition, FMRP associates with neuronal polyribosomes as well as with Staufen-containing ribonucleoprotein (mRNP) granules easily observed in neurites of cultured neurons (Feng et al. 1997; Krichevsky and Kosik 2001; Mazroui et al. 2002; Kanai et al. 2004; Barbee et al. 2006; Bramham and Wells 2007; Bassell and Warren 2008; Dictenberg et al. 2008). FMRP-containing neuronal mRNPs contain not only several ubiquitous translational control molecules, but also CaMKII and Arc mRNAs, whose translation is locally controlled at synapses (Rook et al. 2000; Krichevsky and Kosik 2001; Kanai et al. 2004; Barbee et al. 2006). Thus, FMRP-containing RNA particles are probably translationally repressed and transported along microtubules from the neuronal cell body to synaptic sites in dendrites where local synaptic activity can induce their translation (Kiebler and Bassell 2006; Dictenberg et al. 2008).The functions of FMRP/dFMR1 in mRNA localization as well as miRNA-dependent and independent forms of translational control is likely to require several other regulatory proteins. To identify such proteins, we used a previously designed and validated genetic screen (Wan et al. 2000; Jin et al. 2004a; Zarnescu et al. 2005). The overexpression of dFMR1 in the fly eye causes a “rough-eye” phenotype through a mechanism that requires (a) key residues in dFMR1 that mediate translational repression in vitro; (b) Ago1, a known components of the miRNA pathway; and (c) a DEAD-box helicase called Me31B, which is a highly conserved protein from yeast (Dhh1p) to humans (Rck54/DDX6) functioning in translational repression and present on neuritic mRNPs (Wan et al. 2000; Laggerbauer et al. 2001; Jin et al. 2004a; Coller and Parker 2005; Barbee et al. 2006; Chu and Rana 2006). To identify other Me31B-like translational repressors and neuronal granule components, we screened mutations in 43 candidate proteins for their ability to modify dFMR1 induced rough-eye phenotype. We describe the results of this genetic screen and follow up experiments to address the potential cellular functions of five genes identified as suppressors of sev-dfmr1.  相似文献   

15.
The Nuclear Component of a Cytonuclear Hybrid Incompatibility in Mimulus Maps to a Cluster of Pentatricopeptide Repeat Genes     
Camille M. Barr  Lila Fishman 《Genetics》2010,184(2):455-465
  相似文献   

16.
Unraveling the Complex Trait of Crop Yield With Quantitative Trait Loci Mapping in Brassica napus   总被引:1,自引:0,他引:1  
Jiaqin Shi  Ruiyuan Li  Dan Qiu  Congcong Jiang  Yan Long  Colin Morgan  Ian Bancroft  Jianyi Zhao  Jinling Meng 《Genetics》2009,182(3):851-861
Yield is the most important and complex trait for the genetic improvement of crops. Although much research into the genetic basis of yield and yield-associated traits has been reported, in each such experiment the genetic architecture and determinants of yield have remained ambiguous. One of the most intractable problems is the interaction between genes and the environment. We identified 85 quantitative trait loci (QTL) for seed yield along with 785 QTL for eight yield-associated traits, from 10 natural environments and two related populations of rapeseed. A trait-by-trait meta-analysis revealed 401 consensus QTL, of which 82.5% were clustered and integrated into 111 pleiotropic unique QTL by meta-analysis, 47 of which were relevant for seed yield. The complexity of the genetic architecture of yield was demonstrated, illustrating the pleiotropy, synthesis, variability, and plasticity of yield QTL. The idea of estimating indicator QTL for yield QTL and identifying potential candidate genes for yield provides an advance in methodology for complex traits.YIELD is the most important and complex trait in crops. It reflects the interaction of the environment with all growth and development processes that occur throughout the life cycle (Quarrie et al. 2006). Crop yield is directly and multiply determined by yield-component traits (such as seed weight and seed number). Yield-related traits (such as biomass, harvest index, plant architecture, adaptation, resistance to biotic and abiotic constraints) may also indirectly affect yield by affecting the yield-component traits or by other, unknown mechanisms. Increasing evidence suggests that “fine-mapped” quantitative trait loci (QTL) or genes identified as affecting crop yield involve diverse pathways, such as seed number (Ashikari et al. 2005; Tian et al. 2006b; Burstin et al. 2007; Xie et al. 2008; Xing et al. 2008; Xue et al. 2008), seed weight (Ishimaru 2003; Song et al. 2005; Shomura et al. 2008; Wang et al. 2008; Xie et al. 2006, 2008; Xing et al. 2008; Xue et al. 2008), flowering time (Cockram et al. 2007; Song et al. 2007; Xie et al. 2008; Xue et al. 2008), plant height (Salamini 2003; Ashikari et al. 2005; Xie et al. 2008; Xue et al. 2008), branching (Clark et al. 2006; Burstin et al. 2007; Xing et al. 2008), biomass yield (Quarrie et al. 2006; Burstin et al. 2007), resistance and tolerance to biotic and abiotic stresses (Khush 2001; Brown 2002; Yuan et al. 2002; Waller et al. 2005; Zhang 2007; Warrington et al. 2008), and root architecture (Hochholdinger et al. 2008).Many experiments have explored the genetic basis of yield and yield-associated traits (yield components and yield-related traits) in crops. Summaries of identified QTL have been published for wheat (MacCaferri et al. 2008), barley (Von Korff et al. 2008), rice, and maize (http://www.gramene.org/). The results show several common patterns. First, QTL for yield and yield-associated traits tend to be clustered in the genome, which suggests that the QTL of the yield-associated traits have pleiotropic effects on yield. Second, this kind of pleiotropy has not been well analyzed genetically. The QTL for yield (complicated factor), therefore, have not been associated with any yield-associated traits (relatively simple factors, such as plant height). Therefore, they are unlikely to predict accurately potential candidate genes for yield. Third, only a few loci (rarely >10) have been found for each of these traits. Thus, the genetic architecture of yield has remained ambiguous. Fourth, trials were carried out in a few environments and how the mode of expression of QTL for these complex traits might respond in different environments is unclear.In this study, the genetic architecture of crop yield was analyzed through the QTL mapping of seed yield and eight yield-associated traits in two related populations of rapeseed (Brassica napus) that were grown in 10 natural environments. The complexity of the genetic architecture of seed yield was demonstrated by QTL meta-analysis. The idea of estimating indicator QTL (QTL of yield-associated traits, which are defined as the potential genetic determinants of the colocalized QTL for yield) for yield QTL in conjunction with the identification of candidate genes is described.  相似文献   

17.
The Retinal Determination Gene eyes absent Is Regulated by the EGF Receptor Pathway Throughout Development in Drosophila     
Claire L. Salzer  Yair Elias  Justin P. Kumar 《Genetics》2010,184(1):185-197
  相似文献   

18.
Regulatory Divergence in Drosophila melanogaster and D. simulans,a Genomewide Analysis of Allele-Specific Expression          下载免费PDF全文
Rita M. Graze  Lauren M. McIntyre  Bradley J. Main  Marta L. Wayne  Sergey V. Nuzhdin 《Genetics》2009,183(2):547-561
  相似文献   

19.
Profiling Sex-Specific piRNAs in Zebrafish     
Xiang Zhou  Zhixiang Zuo  Fang Zhou  Wei Zhao  Yuriko Sakaguchi  Takeo Suzuki  Tsutomu Suzuki  Hanhua Cheng  Rongjia Zhou 《Genetics》2010,186(4):1175-1185
Piwi proteins and their partner small RNAs play an essential role in fertility, germ-line stem cell development, and the basic control and evolution of animal genomes. However, little knowledge exists regarding piRNA biogenesis. Utilizing microfluidic chip analysis, we present a quantitative profile of zebrafish piRNAs expressed differentially between testis and ovary. The sex-specific piRNAs are derived from separate loci of repeat elements in the genome. Ovarian piRNAs can be categorized into groups that reach up to 92 members, indicating a sex-specific arrangement of piRNA genes in the genome. Furthermore, precursor piRNAs preferentially form a hairpin structure at the 3′end, which seem to favor the generation of mature sex-specific piRNAs. In addition, the mature piRNAs from both the testis and the ovary are 2′-O-methylated at their 3′ ends.SMALL RNAs, ranging from 19 to 30 nucleotides (nt) in length, constitute a large family of regulatory molecules with diverse functions in invertebrates, vertebrates, plants, and fungi (Bartel 2004; Nakayashiki 2005). Two major classes of small RNAs are microRNAs (miRNAs) and small interfering RNAs (siRNAs). The functions of small RNAs have been conserved through evolution; they have been shown to inhibit gene expression at the levels of mRNA degradation, translational repression, chromatin modification, heterochromatin formation, and DNA elimination (Mochizuki et al. 2002; Bartel 2004; Kim et al. 2005; Brodersen and Voinnet 2006; Lee and Collins 2006; Vaucheret 2006).Over the past few years, focus on the genetics of small RNAs has helped clarify the mechanisms behind the regulation of these molecules. While hundreds of small RNAs have been identified from mammalian somatic tissues, relatively little is known about small RNAs in germ cells. A recent breakthrough has been the identification of small RNAs that associate with Piwi proteins (piRNAs) from Drosophila and mammalian gonads (Aravin et al. 2001, 2006; Girard et al. 2006; Grivna et al. 2006; Vagin et al. 2006; Watanabe et al. 2006). piRNAs and their interacting proteins Ziwi/Zili have also been identified in zebrafish (Houwing et al. 2007, 2008). Increasing evidence indicates that piRNAs play roles mainly in germ cell differentiation and genomic stability (Carthew 2006; Lau et al. 2006; Vagin et al. 2006; Brennecke et al. 2007; Chambeyron et al. 2008; Klattenhoff and Theurkauf 2008; Kuramochi-Miyagawa et al. 2008; Kim et al. 2009; Lim et al. 2009; Unhavaithaya et al. 2009). Moreover, although piRNAs are mostly expressed in germ line cells, recent studies showed piRNA expression in nongerm cells, for example, T-cell lines (Jurkat cells and MT4) (Azuma-Mukai et al. 2008; Yeung et al. 2009), indicating other functions such as in the immune system. piRNAs do not appear to be derived from double-stranded RNA precursors, and their biogenesis mechanisms, although unclear, may be distinct from those of siRNA and miRNA. Recently, two distinct piRNA production pathways were further proposed: the “ping-pong” model (Brennecke et al. 2007; Gunawardane et al. 2007) and the Ago3-independent piRNA pathway centered on Piwi in somatic cells (Li et al. 2009; Malone et al. 2009). However, the mechanistic pathways of piRNA activity and their biogenesis are still largely unknown.Teleost fishes comprise >24,000 species, accounting for more than half of extant vertebrate species, displaying remarkable variation in morphological and physiological adaptations (see review in Zhou et al. 2001). Recently, Houwing et al. (2007, 2008) reported findings on Ziwi/Zili and associated piRNAs, implicating roles in germ cell differentiation, meiosis, and transposon silencing in the germline of the zebrafish. However, some of the identified zebrafish piRNAs are nonrepetitive and nontransposon-related piRNAs, suggesting that piRNAs may have additional unknown roles. In this study, we show that for males and females, piRNAs are specifically derived from separate loci of the repeat elements, and that ovarian piRNAs are far more often associated in groups. Genomic analysis of piRNAs indicates a tendency to folding at the 3′ end of the piRNA precursor, which may favor cleavage of the piRNA precursor to generate mature sex-specific piRNAs. Furthermore, methylation modification occurs at the 2′-O-hydroxyl group on the ribose of the final 3′ nucleotide in both the testis and the ovary.  相似文献   

20.
Patterns and Processes of Genome-Wide Divergence Between North American and African Drosophila melanogaster     
Roman Yukilevich  Thomas L. Turner  Fumio Aoki  Sergey V. Nuzhdin  John R. True 《Genetics》2010,186(1):219-239
Genomic tools and analyses are now being widely used to understand genome-wide patterns and processes associated with speciation and adaptation. In this article, we apply a genomics approach to the model organism Drosophila melanogaster. This species originated in Africa and subsequently spread and adapted to temperate environments of Eurasia and the New World, leading some populations to evolve reproductive isolation, especially between cosmopolitan and Zimbabwean populations. We used tiling arrays to identify highly differentiated regions within and between North America (the United States and Caribbean) and Africa (Cameroon and Zimbabwe) across 63% of the D. melanogaster genome and then sequenced representative fragments to study their genetic divergence. Consistent with previous findings, our results showed that most differentiation was between populations living in Africa vs. outside of Africa (i.e., “out-of-Africa” divergence), with all other geographic differences being less substantial (e.g., between cosmopolitan and Zimbabwean races). The X chromosome was much more strongly differentiated than the autosomes between North American and African populations (i.e., greater X divergence). Overall differentiation was positively associated with recombination rates across chromosomes, with a sharp reduction in regions near centromeres. Fragments surrounding these high FST sites showed reduced haplotype diversity and increased frequency of rare and derived alleles in North American populations compared to African populations. Nevertheless, despite sharp deviation from neutrality in North American strains, a small set of bottleneck/expansion demographic models was consistent with patterns of variation at the majority of our high FST fragments. Although North American populations were more genetically variable compared to Europe, our simulation results were generally consistent with those previously based on European samples. These findings support the hypothesis that most differentiation between North America and Africa was likely driven by the sorting of African standing genetic variation into the New World via Europe. Finally, a few exceptional loci were identified, highlighting the need to use an appropriate demographic null model to identify possible cases of selective sweeps in species with complex demographic histories.THE study of genetic differentiation between populations and species has recently been empowered by the use of genomic techniques and analysis (e.g., Noor and Feder 2006; Stinchcombe and Hoekstra 2008). In the past decade, genetic studies of adaptation and speciation have taken advantage of emerging molecular techniques to scan the genomes of diverging populations for highly differentiated genetic regions (e.g., Wilding et al. 2001; Emelianov et al. 2003; Beaumont and Balding 2004; Campbell and Bernatchez 2004; Scotti-Saintagne et al. 2004; Achere et al. 2005; Turner et al. 2005; Vasemagi et al. 2005; Bonin et al. 2006, 2007; Murray and Hare 2006; Savolainen et al. 2006; Yatabe et al. 2007; Nosil et al. 2008, 2009; Turner et al. 2008a,b; Kulathinal et al. 2009). As a result, genome scans can identify candidate regions that may be associated with adaptive evolution between diverging populations and, more broadly, are able to describe genome-wide patterns and processes of population differentiation (Begun et al. 2007; Stinchcombe and Hoekstra 2008).Genome scans in well-studied genetic model species such as Drosophila melanogaster gain particular power because differentiated loci are mapped to a well-annotated genome. Moreover, the evolutionary history of D. melanogaster is rich with adaptive and demographic events with many parallels to human evolution. Most notable is the historical out-of-Africa migration and subsequent adaptation to temperate ecological environments of Europe, Asia, North America, and Australia. This has resulted in widespread genetic and phenotypic divergence between African and non-African populations (e.g., David and Capy 1988; Begun and Aquadro 1993; Capy et al. 1994; Colegrave et al. 2000; Rouault et al. 2001; Takahashi et al. 2001; Caracristi and Schlötterer 2003; Baudry et al. 2004; Pool and Aquadro 2006; Schmidt et al. 2008; Yukilevich and True 2008a,b). Further, certain populations in Africa and in the Caribbean vary in their degree of reproductive isolation from populations in more temperate regions (Wu et al. 1995; Hollocher et al. 1997; Yukilevich and True 2008a,b). In particular, the Zimbabwe and nearby populations of southern Africa are strongly sexually isolated from all other populations, designating them as a distinct behavioral race (Wu et al. 1995).D. melanogaster has received a great deal of attention from the population geneticists in studying patterns of sequence variation across African and non-African populations. Many snapshots have been taken of random microsatellite and SNP variants spread across X and autosomes, and these have generated several important conclusions. Polymorphism patterns in European populations are characterized by reduced levels of nucleotide and haplotype diversity, an excess of high frequency-derived polymorphisms, and elevated levels of linkage disequilibrium relative to African populations (e.g., Begun and Aquadro 1993; Andolfatto 2001; Glinka et al. 2003; Haddrill et al. 2005; Ometto et al. 2005; Thornton and Andolfatto 2006; Hutter et al. 2007; Singh et al. 2007). These results have been generally interpreted as compatible with population size reduction/bottlenecks followed by recent population expansions. On the other hand, African populations are generally assumed either to have been relatively constant in size over time or to have experienced population size expansions. They generally show higher levels of nucleotide and haplotype diversity, an excess of rare variants, and a deficit of high frequency-derived alleles (Glinka et al. 2003; Ometto et al. 2005; Pool and Aquadro 2006; Hutter et al. 2007; but see Haddrill et al. 2005 for evidence of bottlenecks in Africa).Previous work also shows that the ratio of X-linked to autosomal polymorphism deviates from neutral expectations in opposite directions in African and European populations with more variation on the X than expected in Africa and less variation on the X than expected in Europe (Andolfatto 2001; Kauer et al. 2002; Hutter et al. 2007; Singh et al. 2007). The deviation from neutrality in the ratio of X-autosome polymorphism may be explained by positive selection being more prevalent on the X in Europe and/or by a combination of bottlenecks and male-biased sex ratios in Europe and female-biased sex ratios in Africa (Charlesworth 2001; Hutter et al. 2007; Singh et al. 2007). The selective explanation stems from the argument that, under the hitchhiking selection model, X-linked loci are likely to be more affected by selective sweeps than autosomal loci (Maynard Smith and Haigh 1974; Charlesworth et al. 1987; Vicoso and Charlesworth 2006, 2009).The relative contribution of selective and demographic processes in shaping patterns of genomic variation and differentiation is highly debated (Wall et al. 2002; Glinka et al. 2003; Haddrill et al. 2005; Ometto et al. 2005; Schöfl and Schlötterer 2004; Thornton and Andolfatto 2006; Hutter et al. 2007; Singh et al. 2007; Shapiro et al. 2007; Stephan and Li 2007; Hahn 2008; Macpherson et al. 2008; Noor and Bennett 2009; Sella et al. 2009). This is especially the case in D. melanogaster because derived non-African populations have likely experienced a complex set of demographic events during their migration out of Africa (e.g., Thornton and Andolfatto 2006; Singh et al. 2007; Stephan and Li 2007), making population genetics signatures of demography and selection difficult to tease apart (e.g., Macpherson et al. 2008). Thus it is still unclear what role selection has played in shaping overall patterns of genomic variation and differentiation relative to demographic processes in this species.While there is a long tradition in studying arbitrarily or opportunistically chosen sequences in D. melanogaster, genomic scans that focus particularly on highly differentiated sites across the genome have received much less attention. Such sites are arguably the best candidates to resolve the debate on which processes have shaped genomic differentiation within species (e.g., Przeworski 2002). Recently, a genome-wide scan of cosmopolitan populations in the United States and in Australia was performed to investigate clinal genomic differentiation on the two continents (Turner et al. 2008a). Many single feature polymorphisms differentiating Northern and Southern Hemisphere populations were identified. Among the most differentiated loci in common between continents, 80% were differentiated in the same orientation relative to the Equator, implicating selection as the likely explanation (Turner et al. 2008a). Larger regions of genomic differentiation within and between African and non-African populations have also been discovered, some of them possibly being driven by divergent selection (e.g., Dopman and Hartl 2007; Emerson et al. 2008; Turner et al. 2008a, Aguade 2009). Despite this recent progress, we still know relatively little about large-scale patterns of genomic differentiation in this species, especially between African and non-African populations, and whether most of this differentiation is consistent with demographic processes alone or if it requires selective explanations.In this work, we explicitly focus on identifying differentiated sites across the genome between U.S., Caribbean, West African, and Zimbabwean populations. This allows us to address several fundamental questions related to genomic evolution in D. melanogaster, such as the following: (1) Do genome-wide patterns of differentiation reflect patterns of reproductive isolation? (2) Is genomic differentiation random across and within chromosomes or are some regions overrepresented? (3) What are the population genetics properties of differentiated sites and their surrounding sequences? (4) Can demographic historical processes alone explain most of the observed differentiation on a genome-wide level or is it necessary to involve selection in their explanation?In general, our findings revealed that most genomic differentiation within D. melanogaster shows an out-of-Africa genetic signature. These results are inconsistent with the notion that most genomic differentiation occurs between cosmopolitan and Zimbabwean reproductively isolated races. Further, we found that the X is more differentiated between North American and African populations and more strongly deviates from pure neutrality in North American populations relative to autosomes. Nevertheless, our article shows that much of this deviation from neutrality is broadly consistent with several demographic null models, with a few notable exceptions. Athough this does not exclude selection as a possible alternative mechanism for the observed patterns, it supports the idea that most differentiation in D. melanogaster was likely driven by the sorting of African standing genetic variation into the New World.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号