11 research outputs found

    Fine Haplotype Structure of a Chromosome 17 Region in the Laboratory and Wild Mouse

    No full text
    Extensive linkage disequilibrium among classical laboratory strains represents an obstacle in the high-resolution haplotype mapping of mouse quantitative trait loci (QTL). To determine the potential of wild-derived mouse strains for fine QTL mapping, we constructed a haplotype map of a 250-kb region of the t-complex on chromosome 17 containing the Hybrid sterility 1 (Hst1) gene. We resequenced 33 loci from up to 80 chromosomes of five mouse (sub)species. Trans-species single-nucleotide polymorphisms (SNPs) were rare between Mus m. musculus (Mmmu) and Mus m. domesticus (Mmd). The haplotypes in Mmmu and Mmd differed and therefore strains from these subspecies should not be combined for haplotype-associated mapping. The haplotypes of t-chromosomes differed from all non-t Mmmu and Mmd haplotypes. Half of the SNPs and SN indels but only one of seven longer rearrangements found in classical laboratory strains were useful for haplotype mapping in the wild-derived M. m. domesticus. The largest Mmd haplotype block contained three genes of a highly conserved synteny. The lengths of the haplotype blocks deduced from 36 domesticus chromosomes were in tens of kilobases, suggesting that the wild-derived Mmd strains are suitable for fine interval-specific mapping

    Modulation of Prdm9-controlled meiotic chromosome asynapsis overrides hybrid sterility in mice

    No full text
    Hybrid sterility is one of the reproductive isolation mechanisms leading to speciation. Prdm9, the only known vertebrate hybrid-sterility gene, causes failure of meiotic chromosome synapsis and infertility in male hybrids that are the offspring of two mouse subspecies. Within species, Prdm9 determines the sites of programmed DNA double-strand breaks (DSBs) and meiotic recombination hotspots. To investigate the relation between Prdm9-controlled meiotic arrest and asynapsis, we inserted random stretches of consubspecific homology on several autosomal pairs in sterile hybrids, and analyzed their ability to form synaptonemal complexes and to rescue male fertility. Twenty-seven or more megabases of consubspecific (belonging to the same subspecies) homology fully restored synapsis in a given autosomal pair, and we predicted that two or more DSBs within symmetric hotspots per chromosome are necessary for successful meiosis. We hypothesize that impaired recombination between evolutionarily diverged chromosomes could function as one of the mechanisms of hybrid sterility occurring in various sexually reproducing species

    Mouse consomic strains: Exploiting genetic divergence between Mus m. musculus and Mus m. domesticus subspecies

    No full text
    Consomic (chromosome substitution) strains (CSs) represent the most recent addition to the mouse genetic resources aimed to geneticaly analyze complex trait loci (QTLs). In this study, we report the development of a set of 28 mouse intersubspecific CSs. In each CS, we replaced a single chromosome of the C57BL/6J (B6) inbred strain (mostly Mus m. domesticus) with its homolog from the PWD/Ph inbred strain of the Mus m. musculus subspecies. These two progenitor subspecies diverged less than 1 million years ago and accumulated a large number of genetic differences that constitute a rich resource of genetic variation for QTL analyses. Altogether, the 18 consomic, nine subconsomic, and one conplastic strain covered all 19 autosomes, X and Y sex chromosomes, and mitochondrial DNA. Most CSs had significantly lower reproductive fitness compared with the progenitor strains. CSs homosomic for chromosomes 10 and 11, and the C57BL/6J-Chr X males, failed to reproduce and were substituted by less affected subconsomics carrying either a proximal, central, or distal part of the respective chromosome. A genome-wide scan of 965 DNA markers revealed 99.87% purity of the B6 genetic background. Thirty-three nonsynonymous substitutions were uncovered in the protein-coding regions of the mitochondrial DNA of the B6.PWD-mt conplastic strain. A pilot-phenotyping experiment project revealed a high number of variations among B6.PWD consomics

    Data from: Hybrid sterility locus on Chromosome X controls meiotic recombination rate in mouse

    No full text
    Meiotic recombination safeguards proper segregation of homologous chromosomes into gametes, affects genetic variation within species, and contributes to meiotic chromosome recognition, pairing and synapsis. The Prdm9 gene has a dual role, it controls meiotic recombination by determining the genomic position of crossover hotspots and, in infertile hybrids of house mouse subspecies Mus m. musculus (Mmm) and Mus m. domesticus (Mmd), it further functions as the major hybrid sterility gene. In the latter role Prdm9 interacts with the hybrid sterility X 2 (Hstx2) genomic locus on Chromosome X (Chr X) by a still unknown mechanism. Here we investigated the meiotic recombination rate at the genome-wide level and its possible relation to hybrid sterility. Using immunofluorescence microscopy we quantified the foci of MLH1 DNA mismatch repair protein, the cytological counterparts of reciprocal crossovers, in a panel of inter-subspecific chromosome substitution strains. Two autosomes, Chr 7 and Chr 11, significantly modified the meiotic recombination rate, yet the strongest modifier, designated meiotic recombination 1, Meir1, emerged in the 4.7 Mb Hstx2 genomic locus on Chr X. The male-limited transgressive effect of Meir1 on recombination rate parallels the male-limited transgressive role of Hstx2 in hybrid male sterility. Thus, both genetic factors, the Prdm9 gene and the Hstx2/Meir1 genomic locus, indicate a link between meiotic recombination and hybrid sterility. A strong female-specific modifier of meiotic recombination rate with the effect opposite to Meir1 was localized on Chr X, distally to Meir1. Mapping Meir1 to a narrow candidate interval on Chr X is an important first step towards positional cloning of the respective gene(s) responsible for variation in the global recombination rate between closely related mouse subspecies

    Balcova - raw data

    No full text
    The file structure is the same for all of .csv files. Each column represents one individiual mouse of the genotype stated in the header. Each cell of the column represents MLH1 or RAD51/DMC1 (according to the file name) count of analyzed cell of the given mouse. Strain abbreviation: D# is B6.PWD-Chr#; D7F1 is B6.PWD-Chr7 x B6 F1 The data were analyzed in R 3.2.2 and using its packages as is declared in Materials and Methods of the published paper

    Fine mapping of meiotic recombination 1, <i>Meir1</i>, on Chr X<sup>PWD</sup> using subconsomic strains.

    No full text
    <p>(<b>A</b>) The 4.7 Mb interval of the proximal part of Chr X shared by B6.PWD-Chr X.1s and B6.PWD-Chr X.2, but absent in B6.PWD-Chr X.1 and B6.PWD-Chr X.3, harbors a transgressive modifier of meiotic recombination rate <i>Meir1</i>. The Y axis shows the distance from the centromere in megabases (GRCm38). Chromosome intervals carrying the B6 DNA sequence are depicted in orange, the PWD sequence in blue. (<b>B</b>) The B6.PWD-Chr X.# subconsomics are abbreviated DX.# in column scatters. The significance of differences between strains (excluding PWD) is marked only when p<0.05. See <a href="http://www.plosgenetics.org/article/info:doi/10.1371/journal.pgen.1005906#pgen.1005906.g002" target="_blank">Fig 2</a> for legend.</p

    Meiotic recombination rate in males heterozygous for null mutation or carrying extra copies of <i>Prdm9</i>.

    No full text
    <p>The mean number of MLH1 foci depends on the genetic background of B6 and PWD males but does not reflect differences in the <i>Prdm9</i> copy number. See <a href="http://www.plosgenetics.org/article/info:doi/10.1371/journal.pgen.1005906#pgen.1005906.g002" target="_blank">Fig 2</a> for legend.</p

    Variation in recombination rate of B6 and PWD male mice.

    No full text
    <p>(<b>A</b>) Pachytene spread of a B6 male meiosis shows central elements of synaptonemal complexes of 19 autosomes immunostained for SYCP1 (green), 22 MLH1 foci (red) and foci of centromeric proteins (violet). The number of MLH1 foci per nucleus was used as an equivalent of number of crossovers. The pseudoautosomal region (PAR) of Chr X and Chr Y carries an additional CO mark. (<b>B</b>) Variation of the number of MLH1 foci per pachytene nucleus of PWD and B6 males. The PAR-associated MLH1 foci were not counted. Each dot represents the MLH1 count of one pachytene spermatocyte. Vertical bars designate SD of individual males, horizontal bars represent the mean number of MLH1 foci per analyzed animal. Average MLH1 counts significantly differ between both strains.</p
    corecore