Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Nucleic Acids Res ; 51(5): 2319-2332, 2023 03 21.
Article in English | MEDLINE | ID: mdl-36806949

ABSTRACT

During mammalian spermatogenesis, the paternal genome is extensively remodelled via replacement of histones with protamines forming the highly compact mature sperm nucleus. Compaction occurs in post-meiotic spermatids and is accompanied by extensive double strand break (DSB) formation. We investigate the epigenomic and genomic context of mouse spermatid DSBs, identifying primary sequence motifs, secondary DNA structures and chromatin contexts associated with this damage. Consistent with previously published results we find spermatid DSBs positively associated with short tandem repeats and LINE elements. We further show spermatid DSBs preferentially occur in association with (CA)n, (NA)n and (RY)n repeats, in predicted Z-DNA, are not associated with G-quadruplexes, are preferentially found in regions of low histone mark coverage and engage the remodelling/NHEJ factor BRD4. Locations incurring DSBs in spermatids also show distinct epigenetic profiles throughout later developmental stages: regions retaining histones in mature sperm, regions susceptible to oxidative damage in mature sperm, and fragile two-cell like embryonic stem cell regions bound by ZSCAN4 all co-localise with spermatid DSBs and with each other. Our results point to a common 'vulnerability code' unifying several types of DNA damage occurring on the paternal genome during reproduction, potentially underpinned by torsional changes during sperm chromatin remodelling.


Subject(s)
Histones , Nuclear Proteins , Male , Mice , Animals , Histones/genetics , Histones/metabolism , Nuclear Proteins/metabolism , Semen/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Chromatin/genetics , Chromatin/metabolism , Spermatids/metabolism , Spermatogenesis/genetics , DNA Damage , Mammals/genetics
2.
PLoS Genet ; 15(7): e1008290, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31329581

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pgen.1002900.].

3.
Reproduction ; 159(4): X1, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32065737

ABSTRACT

The journal and the authors apologise for an error in the above titled article published in this journal (vol 144, pp 433­445). The authors inadvertently presented duplicate sperm images for XY and XESxrbO mouse testes of Fig. 6 (bottom panels). This error does not change the findings of the paper, as this figure does not give a quantitative breakdown of the proportions of different shapes.

4.
Genome Res ; 26(1): 130-9, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26560630

ABSTRACT

We have generated an improved assembly and gene annotation of the pig X Chromosome, and a first draft assembly of the pig Y Chromosome, by sequencing BAC and fosmid clones from Duroc animals and incorporating information from optical mapping and fiber-FISH. The X Chromosome carries 1033 annotated genes, 690 of which are protein coding. Gene order closely matches that found in primates (including humans) and carnivores (including cats and dogs), which is inferred to be ancestral. Nevertheless, several protein-coding genes present on the human X Chromosome were absent from the pig, and 38 pig-specific X-chromosomal genes were annotated, 22 of which were olfactory receptors. The pig Y-specific Chromosome sequence generated here comprises 30 megabases (Mb). A 15-Mb subset of this sequence was assembled, revealing two clusters of male-specific low copy number genes, separated by an ampliconic region including the HSFY gene family, which together make up most of the short arm. Both clusters contain palindromes with high sequence identity, presumably maintained by gene conversion. Many of the ancestral X-related genes previously reported in at least one mammalian Y Chromosome are represented either as active genes or partial sequences. This sequencing project has allowed us to identify genes--both single copy and amplified--on the pig Y Chromosome, to compare the pig X and Y Chromosomes for homologous sequences, and thereby to reveal mechanisms underlying pig X and Y Chromosome evolution.


Subject(s)
Chromosomes, Mammalian/genetics , Evolution, Molecular , Swine/genetics , X Chromosome/genetics , Y Chromosome/genetics , Animals , Base Sequence , Cats/genetics , Dogs/genetics , Female , Gene Conversion , Gene Expression , Gene Library , Gene Order , Humans , Male , Molecular Sequence Data , Sequence Alignment , Sequence Analysis, DNA
5.
Hum Mol Genet ; 25(24): 5300-5310, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27742779

ABSTRACT

During spermatogenesis, germ cells that fail to synapse their chromosomes or fail to undergo meiotic sex chromosome inactivation (MSCI) are eliminated via apoptosis during mid-pachytene. Previous work showed that Y-linked genes Zfy1 and Zfy2 act as 'executioners' for this checkpoint, and that wrongful expression of either gene during pachytene triggers germ cell death. Here, we show that in mice, Zfy genes are also necessary for efficient MSCI and the sex chromosomes are not correctly silenced in Zfy-deficient spermatocytes. This unexpectedly reveals a triple role for Zfy at the mid-pachytene checkpoint in which Zfy genes first promote MSCI, then monitor its progress (since if MSCI is achieved, Zfy genes will be silenced), and finally execute cells with MSCI failure. This potentially constitutes a negative feedback loop governing this critical checkpoint mechanism.


Subject(s)
DNA-Binding Proteins/genetics , Spermatocytes/metabolism , Transcription Factors/genetics , X Chromosome Inactivation/genetics , Animals , Male , Meiosis/genetics , Mice , Spermatocytes/growth & development , Spermatogenesis/genetics , X Chromosome/genetics
7.
Development ; 141(4): 855-66, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24496622

ABSTRACT

Outbred XY(Sry-) female mice that lack Sry due to the 11 kb deletion Sry(dl1Rlb) have very limited fertility. However, five lines of outbred XY(d) females with Y chromosome deletions Y(Del(Y)1Ct)-Y(Del(Y)5Ct) that deplete the Rbmy gene cluster and repress Sry transcription were found to be of good fertility. Here we tested our expectation that the difference in fertility between XO, XY(d-1) and XY(Sry-) females would be reflected in different degrees of oocyte depletion, but this was not the case. Transgenic addition of Yp genes to XO females implicated Zfy2 as being responsible for the deleterious Y chromosomal effect on fertility. Zfy2 transcript levels were reduced in ovaries of XY(d-1) compared with XY(Sry-) females in keeping with their differing fertility. In seeking the biological basis of the impaired fertility we found that XY(Sry-), XY(d-1) and XO,Zfy2 females produce equivalent numbers of 2-cell embryos. However, in XY(Sry-) and XO,Zfy2 females the majority of embryos arrested with 2-4 cells and almost no blastocysts were produced; by contrast, XY(d-1) females produced substantially more blastocysts but fewer than XO controls. As previously documented for C57BL/6 inbred XY females, outbred XY(Sry-) and XO,Zfy2 females showed frequent failure of the second meiotic division, although this did not prevent the first cleavage. Oocyte transcriptome analysis revealed major transcriptional changes resulting from the Zfy2 transgene addition. We conclude that Zfy2-induced transcriptional changes in oocytes are sufficient to explain the more severe fertility impairment of XY as compared with XO females.


Subject(s)
DNA-Binding Proteins/metabolism , Infertility, Female/genetics , Meiosis/genetics , Oocytes/metabolism , Sex Chromosome Disorders of Sex Development/genetics , Sex-Determining Region Y Protein/deficiency , Transcription Factors/metabolism , Y Chromosome/genetics , Animals , Blotting, Western , Breeding , Cleavage Stage, Ovum/pathology , Cleavage Stage, Ovum/physiology , Crosses, Genetic , DNA-Binding Proteins/genetics , Female , Gene Expression Profiling , Gene Expression Regulation/genetics , Genotype , Linear Models , Mice , Mice, Transgenic , Microarray Analysis , Transcription Factors/genetics
8.
J Cell Sci ; 126(Pt 3): 803-13, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23178944

ABSTRACT

In mouse and man Y chromosome deletions are frequently associated with spermatogenic defects. Mice with extensive deletions of non-pairing Y chromosome long arm (NPYq) are infertile and produce sperm with grossly misshapen heads, abnormal chromatin packaging and DNA damage. The NPYq-encoded multi-copy gene Sly controls the expression of sex chromosome genes after meiosis and Sly deficiency results in a remarkable upregulation of sex chromosome genes. Sly deficiency has been shown to be the underlying cause of the sperm head anomalies and infertility associated with NPYq gene loss, but it was not known whether it recapitulates sperm DNA damage phenotype. We produced and examined mice with transgenically (RNAi) silenced Sly and demonstrated that these mice have increased incidence of sperm with DNA damage and poorly condensed and insufficiently protaminated chromatin. We also investigated the contribution of each of the two Sly-encoded transcript variants and noted that the phenotype was only observed when both variants were knocked down, and that the phenotype was intermediate in severity compared with mice with severe NPYq deficiency. Our data demonstrate that Sly deficiency is responsible for the sperm DNA damage/chromatin packaging defects observed in mice with NPYq deletions and point to SLY proteins involvement in chromatin reprogramming during spermiogenesis, probably through their effect on the post-meiotic expression of spermiogenic genes. Considering the importance of the sperm epigenome for embryonic and fetal development and the possibility of its inter-generational transmission, our results are important for future investigations of the molecular mechanisms of this biologically and clinically important process.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Nuclear Proteins/metabolism , Spermatozoa/metabolism , Y Chromosome/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Vesicular Transport , Animals , Base Sequence , Cells, Cultured , Chromatin Assembly and Disassembly/genetics , DNA Damage/genetics , Female , Gene Dosage , Humans , Infertility, Male , Male , Mice , Mice, Transgenic , Molecular Sequence Data , Nuclear Proteins/genetics , RNA, Small Interfering/genetics , Sequence Deletion/genetics , Transgenes/genetics
9.
PLoS Genet ; 8(9): e1002900, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23028340

ABSTRACT

Intragenomic conflicts arise when a genetic element favours its own transmission to the detriment of others. Conflicts over sex chromosome transmission are expected to have influenced genome structure, gene regulation, and speciation. In the mouse, the existence of an intragenomic conflict between X- and Y-linked multicopy genes has long been suggested but never demonstrated. The Y-encoded multicopy gene Sly has been shown to have a predominant role in the epigenetic repression of post meiotic sex chromatin (PMSC) and, as such, represses X and Y genes, among which are its X-linked homologs Slx and Slxl1. Here, we produced mice that are deficient for both Sly and Slx/Slxl1 and observed that Slx/Slxl1 has an opposite role to that of Sly, in that it stimulates XY gene expression in spermatids. Slx/Slxl1 deficiency rescues the sperm differentiation defects and near sterility caused by Sly deficiency and vice versa. Slx/Slxl1 deficiency also causes a sex ratio distortion towards the production of male offspring that is corrected by Sly deficiency. All in all, our data show that Slx/Slxl1 and Sly have antagonistic effects during sperm differentiation and are involved in a postmeiotic intragenomic conflict that causes segregation distortion and male sterility. This is undoubtedly what drove the massive gene amplification on the mouse X and Y chromosomes. It may also be at the basis of cases of F1 male hybrid sterility where the balance between Slx/Slxl1 and Sly copy number, and therefore expression, is disrupted. To the best of our knowledge, our work is the first demonstration of a competition occurring between X and Y related genes in mammals. It also provides a biological basis for the concept that intragenomic conflict is an important evolutionary force which impacts on gene expression, genome structure, and speciation.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Nuclear Proteins/genetics , Proteins/genetics , X Chromosome/genetics , Y Chromosome/genetics , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Vesicular Transport , Animals , Epigenesis, Genetic , Female , Gene Dosage , Gene Expression Regulation , Genetic Speciation , Infertility, Male , Male , Meiosis/genetics , Mice , Mice, Transgenic , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/deficiency , Proteins/antagonists & inhibitors , Sex Chromatin/genetics , Sex Chromatin/metabolism , Sex Ratio , Spermatids/metabolism , Spermatozoa/growth & development , Spermatozoa/metabolism
10.
BMC Genomics ; 15: 49, 2014 Jan 21.
Article in English | MEDLINE | ID: mdl-24447410

ABSTRACT

BACKGROUND: Maternal undernutrition leads to an increased risk of metabolic disorders in offspring including obesity and insulin resistance, thought to be due to a programmed thrifty phenotype which is inappropriate for a subsequent richer nutritional environment. In a rat model, both male and female offspring of undernourished mothers are programmed to become obese, however postnatal leptin treatment gives discordant results between males and females. Leptin treatment is able to rescue the adverse programming effects in the female offspring of undernourished mothers, but not in their male offspring. Additionally, in these rats, postnatal leptin treatment of offspring from normally-nourished mothers programmes their male offspring to develop obesity in later life, while there is no comparable effect in their female offspring. RESULTS: We show by microarray analysis of the female liver transcriptome that both maternal undernutrition and postnatal leptin treatment independently induce a similar thrifty transcriptional programme affecting carbohydrate metabolism, amino acid metabolism and oxidative stress genes. Paradoxically, however, the combination of both stimuli restores a more normal transcriptional environment. This demonstrates that "leptin reversal" is a global phenomenon affecting all genes involved in fetal programming by maternal undernourishment and leptin treatment. The thrifty transcriptional programme was associated with pro-inflammatory markers and downregulation of adaptive immune mediators, particularly MHC class I genes, suggesting a deficit in antigen presentation in these offspring. CONCLUSIONS: We propose a revised model of developmental programming reconciling the male and female observations, in which there are two competing programmes which collectively drive liver transcription. The first element is a thrifty metabolic phenotype induced by early life growth restriction independently of leptin levels. The second is a homeostatic set point calibrated in response to postnatal leptin surge, which is able to over-ride the metabolic programme. This "calibration model" for the postnatal leptin surge, if applicable in humans, may have implications for understanding responses to catch-up growth in infants. Additionally, the identification of an antigen presentation deficit associated with metabolic thriftiness may relate to a previously observed correlation between birth season (a proxy for gestational undernutrition) and infectious disease mortality in rural African communities.


Subject(s)
Fetal Nutrition Disorders/genetics , Leptin/pharmacology , Liver/drug effects , Amino Acids/metabolism , Animals , Carbohydrate Metabolism/genetics , Diet , Disease Models, Animal , Female , Fetal Development , Fetal Nutrition Disorders/metabolism , Fetal Nutrition Disorders/pathology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Inflammation Mediators/metabolism , Liver/metabolism , Male , Obesity/metabolism , Obesity/pathology , Oxidative Stress/genetics , Phenotype , Pregnancy , Rats , Rats, Wistar , Transcriptome/drug effects
11.
Hum Mol Genet ; 20(15): 3010-21, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21551453

ABSTRACT

In common with other mammalian sex chromosomes, the mouse sex chromosomes are enriched for genes with male-specific function such as testis genes. However, in mouse there has been an unprecedented expansion of ampliconic sequence containing spermatid-expressed genes. We show via a phylogenetic analysis of gene amplification on the mouse sex chromosomes that multiple families of sex-linked spermatid-expressed genes are highly amplified in Mus musculus subspecies and in two further species from the Palaearctic clade of mouse species. Ampliconic X-linked genes expressed in other cell types showed a different evolutionary trajectory, without the distinctive simultaneous amplification seen in spermatid-expressed genes. The Palaearctic gene amplification occurred concurrently with the appearance of Sly, a Yq-linked regulator of post-meiotic sex chromatin (PMSC) which acts to repress sex chromosome transcription in spermatids. Despite the gene amplification, there was comparatively little effect on transcript abundance, suggesting that the genes in question became amplified in order to overcome Sly-mediated transcriptional repression and maintain steady expression levels in spermatids. Together with the known sex-ratio effects of Yq/Sly deficiency, our results suggest that Sly is involved in a genomic conflict with one or more X-linked sex-ratio distorter genes. The recent evolution of the novel PMSC regulator Sly in mouse lineages has significant implications for the use of mouse-model systems in investigating sex chromosome dynamics in spermatids.


Subject(s)
Genes, X-Linked/genetics , Spermatids/metabolism , Animals , Biological Evolution , Gene Amplification/genetics , Male , Mice , Phylogeny , Sex Chromosomes/genetics
12.
Nat Commun ; 14(1): 2855, 2023 05 18.
Article in English | MEDLINE | ID: mdl-37202403

ABSTRACT

NDP52 is an autophagy receptor involved in the recognition and degradation of invading pathogens and damaged organelles. Although NDP52 was first identified in the nucleus and is expressed throughout the cell, to date, there is no clear nuclear functions for NDP52. Here, we use a multidisciplinary approach to characterise the biochemical properties and nuclear roles of NDP52. We find that NDP52 clusters with RNA Polymerase II (RNAPII) at transcription initiation sites and that its overexpression promotes the formation of additional transcriptional clusters. We also show that depletion of NDP52 impacts overall gene expression levels in two model mammalian cells, and that transcription inhibition affects the spatial organisation and molecular dynamics of NDP52 in the nucleus. This directly links NDP52 to a role in RNAPII-dependent transcription. Furthermore, we also show that NDP52 binds specifically and with high affinity to double-stranded DNA (dsDNA) and that this interaction leads to changes in DNA structure in vitro. This, together with our proteomics data indicating enrichment for interactions with nucleosome remodelling proteins and DNA structure regulators, suggests a possible function for NDP52 in chromatin regulation. Overall, here we uncover nuclear roles for NDP52 in gene expression and DNA structure regulation.


Subject(s)
Nuclear Proteins , RNA Polymerase II , Animals , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Nuclear Proteins/metabolism , Autophagy/genetics , DNA/genetics , DNA/metabolism , Nucleic Acid Conformation , Mammals/genetics
13.
Reproduction ; 144(4): 433-45, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22869781

ABSTRACT

We recently used three XO male mouse models with varying Y short-arm (Yp) gene complements, analysed at 30 days post partum, to demonstrate a Yp gene requirement for the apoptotic elimination of spermatocytes with a univalent X chromosome at the first meiotic metaphase. The three mouse models were i) XSxr(a)O in which the Yp-derived Tp(Y)1Ct(Sxr-a) sex reversal factor provides an almost complete Yp gene complement, ii) XSxr(b)O,Eif2s3y males in which Tp(Y)1Ct(Sxr-b) has a deletion completely or partially removing eight Yp genes - the Yp gene Eif2s3y has been added as a transgene to support spermatogonial proliferation, and iii) XOSry,Eif2s3y males in which the Sry transgene directs gonad development along the male pathway. In this study, we have used the same mouse models analysed at 6 weeks of age to investigate potential Yp gene involvement in spermiogenesis. We found that all three mouse models produce haploid and diploid spermatids and that the diploid spermatids showed frequent duplication of the developing acrosomal cap during the early stages. However, only in XSxr(a)O males did spermiogenesis continue to completion. Most strikingly, in XOSry,Eif2s3y males, spermatid development arrested at round spermatid step 7 so that no sperm head restructuring or tail development was observed. In contrast, in XSxr(b)O,Eif2s3y males, spermatids with substantial sperm head and tail morphogenesis could be easily found, although this was delayed compared with XSxr(a)O. We conclude that Sxr(a) (and therefore Yp) includes genetic information essential for sperm morphogenesis and that this is partially retained in Sxr(b).


Subject(s)
Disease Models, Animal , Eukaryotic Initiation Factor-2/metabolism , Genes, Y-Linked , Sex Chromosome Disorders of Sex Development/metabolism , Sex-Determining Region Y Protein/metabolism , Spermatids/metabolism , Spermatogenesis , Acrosome/metabolism , Acrosome/pathology , Animals , Chromosome Deletion , Chromosomes, Human, Y/metabolism , Crosses, Genetic , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Eukaryotic Initiation Factor-2/genetics , Gene Deletion , Infertility, Male , Male , Meiosis , Mice , Mice, Knockout , Mice, Transgenic , Recombinant Fusion Proteins/metabolism , Sex Chromosome Aberrations , Sex Chromosome Disorders of Sex Development/pathology , Sex-Determining Region Y Protein/genetics , Sperm Tail/metabolism , Sperm Tail/pathology , Spermatids/pathology , Transcription Factors/genetics , Transcription Factors/metabolism
14.
PLoS Biol ; 7(11): e1000244, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19918361

ABSTRACT

Studies of mice with Y chromosome long arm deficiencies suggest that the male-specific region (MSYq) encodes information required for sperm differentiation and postmeiotic sex chromatin repression (PSCR). Several genes have been identified on MSYq, but because they are present in more than 40 copies each, their functions cannot be investigated using traditional gene targeting. Here, we generate transgenic mice producing small interfering RNAs that specifically target the transcripts of the MSYq-encoded multicopy gene Sly (Sycp3-like Y-linked). Microarray analyses performed on these Sly-deficient males and on MSYq-deficient males show a remarkable up-regulation of sex chromosome genes in spermatids. SLY protein colocalizes with the X and Y chromatin in spermatids of normal males, and Sly deficiency leads to defective repressive marks on the sex chromatin, such as reduced levels of the heterochromatin protein CBX1 and of histone H3 methylated at lysine 9. Sly-deficient mice, just like MSYq-deficient mice, have severe impairment of sperm differentiation and are near sterile. We propose that their spermiogenesis phenotype is a consequence of the change in spermatid gene expression following Sly deficiency. To our knowledge, this is the first successful targeted disruption of the function of a multicopy gene (or of any Y gene). It shows that SLY has a predominant role in PSCR, either via direct interaction with the spermatid sex chromatin or via interaction with sex chromatin protein partners. Sly deficiency is the major underlying cause of the spectrum of anomalies identified 17 y ago in MSYq-deficient males. Our results also suggest that the expansion of sex-linked spermatid-expressed genes in mouse is a consequence of the enhancement of PSCR that accompanies Sly amplification.


Subject(s)
Gene Dosage , Germ Cells/cytology , Meiosis , Y Chromosome , Animals , Chromosomes, Mammalian , Gene Expression Regulation , Infertility, Male/genetics , Male , Mice , Mice, Transgenic , Sex Chromosomes , Spermatids
15.
Genetics ; 222(4)2022 11 30.
Article in English | MEDLINE | ID: mdl-36194004

ABSTRACT

Incompatibilities on the sex chromosomes are important in the evolution of hybrid male sterility, but the evolutionary forces underlying this phenomenon are unclear. House mice (Mus musculus) lineages have provided powerful models for understanding the genetic basis of hybrid male sterility. X chromosome-autosome interactions cause strong incompatibilities in M. musculus F1 hybrids, but variation in sterility phenotypes suggests a more complex genetic basis. In addition, XY chromosome conflict has resulted in rapid expansions of ampliconic genes with dosage-dependent expression that is essential to spermatogenesis. Here, we evaluated the contribution of XY lineage mismatch to male fertility and stage-specific gene expression in hybrid mice. We performed backcrosses between two house mouse subspecies to generate reciprocal Y-introgression strains and used these strains to test the effects of XY mismatch in hybrids. Our transcriptome analyses of sorted spermatid cells revealed widespread overexpression of the X chromosome in sterile F1 hybrids independent of Y chromosome subspecies origin. Thus, postmeiotic overexpression of the X chromosome in sterile F1 mouse hybrids is likely a downstream consequence of disrupted meiotic X-inactivation rather than XY gene copy number imbalance. Y chromosome introgression did result in subfertility phenotypes and disrupted expression of several autosomal genes in mice with an otherwise nonhybrid genomic background, suggesting that Y-linked incompatibilities contribute to reproductive barriers, but likely not as a direct consequence of XY conflict. Collectively, these findings suggest that rapid sex chromosome gene family evolution driven by genomic conflict has not resulted in strong male reproductive barriers between these subspecies of house mice.


Subject(s)
Hybridization, Genetic , Infertility, Male , Humans , Male , Mice , Animals , Spermatogenesis/genetics , Sex Chromosomes/genetics , X Chromosome/genetics , Infertility, Male/genetics
16.
Nat Commun ; 13(1): 2608, 2022 05 11.
Article in English | MEDLINE | ID: mdl-35546158

ABSTRACT

Chromosome folding has profound impacts on gene regulation, whose evolutionary consequences are far from being understood. Here we explore the relationship between 3D chromatin remodelling in mouse germ cells and evolutionary changes in genome structure. Using a comprehensive integrative computational analysis, we (i) reconstruct seven ancestral rodent genomes analysing whole-genome sequences of 14 species representatives of the major phylogroups, (ii) detect lineage-specific chromosome rearrangements and (iii) identify the dynamics of the structural and epigenetic properties of evolutionary breakpoint regions (EBRs) throughout mouse spermatogenesis. Our results show that EBRs are devoid of programmed meiotic DNA double-strand breaks (DSBs) and meiotic cohesins in primary spermatocytes, but are associated in post-meiotic cells with sites of DNA damage and functional long-range interaction regions that recapitulate ancestral chromosomal configurations. Overall, we propose a model that integrates evolutionary genome reshuffling with DNA damage response mechanisms and the dynamic spatial genome organisation of germ cells.


Subject(s)
Chromatin Assembly and Disassembly , Germ Cells , Animals , Chromatin/genetics , Chromatin Assembly and Disassembly/genetics , DNA Breaks, Double-Stranded , Genome , Male , Meiosis/genetics , Mice , Spermatogenesis/genetics
17.
Nat Commun ; 13(1): 5818, 2022 10 07.
Article in English | MEDLINE | ID: mdl-36207323

ABSTRACT

Human papillomavirus (HPV)-associated cervical cancer is a leading cause of cancer deaths in women. Here we present an integrated multi-omic analysis of 643 cervical squamous cell carcinomas (CSCC, the most common histological variant of cervical cancer), representing patient populations from the USA, Europe and Sub-Saharan Africa and identify two CSCC subtypes (C1 and C2) with differing prognosis. C1 and C2 tumours can be driven by either of the two most common HPV types in cervical cancer (16 and 18) and while HPV16 and HPV18 are overrepresented among C1 and C2 tumours respectively, the prognostic difference between groups is not due to HPV type. C2 tumours, which comprise approximately 20% of CSCCs across these cohorts, display distinct genomic alterations, including loss or mutation of the STK11 tumour suppressor gene, increased expression of several immune checkpoint genes and differences in the tumour immune microenvironment that may explain the shorter survival associated with this group. In conclusion, we identify two therapy-relevant CSCC subtypes that share the same defining characteristics across three geographically diverse cohorts.


Subject(s)
Carcinoma, Squamous Cell , Papillomavirus Infections , Uterine Cervical Neoplasms , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Female , Human papillomavirus 16/genetics , Humans , Papillomaviridae/genetics , Papillomavirus Infections/complications , Papillomavirus Infections/genetics , Papillomavirus Infections/pathology , Prognosis , Tumor Microenvironment , Uterine Cervical Neoplasms/pathology
18.
Dev Cell ; 10(4): 521-9, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16580996

ABSTRACT

Transcriptional silencing of the sex chromosomes during male meiosis (MSCI) is conserved among organisms with limited sex chromosome synapsis, including mammals. Since the 1990s the prevailing view has been that MSCI in mammals is transient, with sex chromosome reactivation occurring as cells exit meiosis. Recently, we found that any chromosome region unsynapsed during pachytene of male and female mouse meiosis is subject to transcriptional silencing (MSUC), and we hypothesized that MSCI is an inevitable consequence of this more general meiotic silencing mechanism. Here, we provide direct evidence that asynapsis does indeed drive MSCI. We also show that a substantial degree of transcriptional repression of the sex chromosomes is retained postmeiotically, and we provide evidence that this postmeiotic repression is a downstream consequence of MSCI/MSUC. While this postmeiotic repression occurs after the loss of MSUC-related proteins at the end of prophase, other histone modifications associated with transcriptional repression have by then become established.


Subject(s)
Meiosis/genetics , Pachytene Stage/genetics , Sex Chromosomes/genetics , Spermatids/physiology , Animals , Female , Gene Silencing , In Situ Hybridization, Fluorescence/methods , Male , Mice , Sex Chromosomes/metabolism , Spermatids/cytology , Transcription, Genetic , X Chromosome/genetics , X Chromosome/metabolism , X Chromosome Inactivation/genetics , XYY Karyotype/genetics , Y Chromosome/genetics , Y Chromosome/metabolism
19.
Reproduction ; 142(5): 609-19, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21911442

ABSTRACT

The ability to pre-select offspring sex via separation of X- and Y-bearing sperm would have profound ramifications for the animal husbandry industry. No fully satisfactory method is as yet available for any species, although flow sorting is commercially viable for cattle. The discovery of antigens that distinguish X- and Y-bearing sperm, i.e. offspring sex-specific antigens (OSSAs), would allow for batched immunological separation of sperm and thus enable a safer, more widely applicable and high-throughput means of sperm sorting. This review addresses the basic processes of spermatogenesis that have complicated the search for OSSAs, in particular the syncytial development of male germ cells, and the transcriptional dynamics of the sex chromosomes during and after meiosis. We survey the various approaches taken to discover OSSA and propose that a whole-genome transcriptional approach to the problem is the most promising avenue for future research in the field.


Subject(s)
Antigens/isolation & purification , Gene Expression Regulation , Sex Chromosomes/genetics , Sex Preselection/methods , Spermatozoa/metabolism , Animals , Antigens/genetics , Antigens/metabolism , Cattle , Female , Gene Expression Profiling , Humans , Kinetics , Male , Models, Biological , Seminal Plasma Proteins/genetics , Seminal Plasma Proteins/immunology , Sex Differentiation/genetics , Sex Preselection/veterinary , Spermatozoa/cytology , Spermatozoa/immunology
SELECTION OF CITATIONS
SEARCH DETAIL