Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Biol Reprod ; 110(5): 985-999, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38376238

ABSTRACT

Sry on the Y-chromosome upregulates Sox9, which in turn upregulates a set of genes such as Fgf9 to initiate testicular differentiation in the XY gonad. In the absence of Sry expression, genes such as Rspo1, Foxl2, and Runx1 support ovarian differentiation in the XX gonad. These two pathways antagonize each other to ensure the development of only one gonadal sex in normal development. In the B6.YTIR mouse, carrying the YTIR-chromosome on the B6 genetic background, Sry is expressed in a comparable manner with that in the B6.XY mouse, yet, only ovaries or ovotestes develop. We asked how testicular and ovarian differentiation pathways interact to determine the gonadal sex in the B6.YTIR mouse. Our results showed that (1) transcript levels of Sox9 were much lower than in B6.XY gonads while those of Rspo1 and Runx1 were as high as B6.XX gonads at 11.5 and 12.5 days postcoitum. (2) FOXL2-positive cells appeared in mosaic with SOX9-positive cells at 12.5 days postcoitum. (3) SOX9-positive cells formed testis cords in the central area while those disappeared to leave only FOXL2-positive cells in the poles or the entire area at 13.5 days postcoitum. (4) No difference was found at transcript levels of all genes between the left and right gonads up to 12.5 days postcoitum, although ovotestes developed much more frequently on the left than the right at 13.5 days postcoitum. These results suggest that inefficient Sox9 upregulation and the absence of Rspo1 repression prevent testicular differentiation in the B6.YTIR gonad.


Subject(s)
SOX9 Transcription Factor , Sex Determination Processes , Testis , Thrombospondins , Up-Regulation , Animals , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Male , Female , Mice , Thrombospondins/genetics , Thrombospondins/metabolism , Sex Determination Processes/genetics , Sex Determination Processes/physiology , Testis/metabolism , Gonads/metabolism , Ovary/metabolism , Forkhead Box Protein L2/genetics , Forkhead Box Protein L2/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Gene Expression Regulation, Developmental , Sex Differentiation/genetics , Mice, Inbred C57BL
2.
Mamm Genome ; 34(1): 44-55, 2023 03.
Article in English | MEDLINE | ID: mdl-36454369

ABSTRACT

Several lines of evidence suggest that the presence of the Y chromosome influences DNA methylation of autosomal loci. To better understand the impact of the Y chromosome on autosomal DNA methylation patterns and its contribution to sex bias in methylation, we identified Y chromosome dependent differentially methylated regions (yDMRs) using whole-genome bisulfite sequencing methylation data from livers of mice with different combinations of sex-chromosome complement and gonadal sex. Nearly 90% of the autosomal yDMRs mapped to transposable elements (TEs) and most of them had lower methylation in XY compared to XX or XO mice. Follow-up analyses of four reporter autosomal yDMRs showed that Y-dependent methylation levels were consistent across most somatic tissues but varied in strains with different origins of the Y chromosome, suggesting that genetic variation in the Y chromosome influenced methylation levels of autosomal regions. Mice lacking the q-arm of the Y chromosome (B6.NPYq-2) as well as mice with a loss-of-function mutation in Kdm5d showed no differences in methylation levels compared to wild type mice. In conclusion, the Y-linked modifier of TE methylation is likely to reside on the short arm of Y chromosome and further studies are required to identify this gene.


Subject(s)
DNA Methylation , Sexism , Mice , Animals , Y Chromosome , Genetic Variation
4.
Sci Rep ; 12(1): 12028, 2022 07 14.
Article in English | MEDLINE | ID: mdl-35835815

ABSTRACT

Meiotic homologous recombination during fetal development dictates proper chromosome segregation in adult mammalian oocytes. Successful homologous synapsis and recombination during Meiotic Prophase I (MPI) depends on telomere-led chromosome movement along the nuclear envelope. In mice, all chromosomes are acrocentric, while other mammalian species carry a mixture of acrocentric and metacentric chromosomes. Such differences in telomeric structures may explain the exceptionally low aneuploidy rates in mice. Here, we tested whether the presence of metacentric chromosomes carrying Robertsonian translocations (RbT) affects the rate of homologous recombination or aneuploidy. We found a delay in MPI progression in RbT-carrier vs. wild-type (WT) fetal ovaries. Furthermore, resolution of distal telomere clusters, associated with synapsis initiation, was delayed and centromeric telomere clusters persisted until later MPI substages in RbT-carrier oocytes compared to WT oocytes. When chromosomes fully synapsed, higher percentages of RbT-carrier oocytes harbored at least one chromosome pair lacking MLH1 foci, which indicate crossover sites, compared to WT oocytes. Aneuploidy rates in ovulated eggs were also higher in RbT-carrier females than in WT females. In conclusion, the presence of metacentric chromosomes among acrocentric chromosomes in mouse oocytes delays MPI progression and reduces the efficiency of homologous crossover, resulting in a higher frequency of aneuploidy.


Subject(s)
Meiosis , Oocytes , Aneuploidy , Animals , Chromosomes , Female , Mammals , Meiosis/genetics , Meiotic Prophase I/genetics , Mice , Telomere/genetics , Translocation, Genetic
5.
Sex Dev ; 16(5-6): 355-364, 2022.
Article in English | MEDLINE | ID: mdl-35235936

ABSTRACT

BACKGROUND: In eutherian mammals, the sex chromosome complement, XX and XY, determines sexual differentiation of gonadal primordia into testes and ovaries, which in turn direct differentiation of germ cells into haploid sperm and oocytes, respectively. When gonadal sex is reversed, however, the germ cell sex becomes discordant with the chromosomal sex. XY females in humans are infertile, while XY females in the mouse (Mus musculus) are subfertile or infertile dependent on the cause of sex reversal and the genetic background. This article reviews publications to understand how the sex chromosome complement affects the fertility of XY oocytes by comparing with XX and monosomy X (XO) oocytes. SUMMARY: The results highlight 2 folds disadvantage of XY oocytes over XX oocytes: (1) the X and Y chromosomes fail to pair during the meiotic prophase I, resulting in sex chromosome aneuploidy at the first meiotic division and (2) expression of the Y-linked genes during oocyte growth affects the transcriptome landscape and renders the ooplasmic component incompetent for embryonic development. KEY MESSAGE: The XX chromosome complement gives the oocyte the highest competence for embryonic development.

7.
Sci Rep ; 11(1): 13766, 2021 07 02.
Article in English | MEDLINE | ID: mdl-34215813

ABSTRACT

Sexual dimorphism in gene regulation, including DNA methylation, is the main driver of sexual dimorphism in phenotypes. However, the questions of how and when sex shapes DNA methylation remain unresolved. Recently, using mice with different combinations of genetic and phenotypic sex, we identified sex-associated differentially methylated regions (sDMRs) that depended on the sex phenotype. Focusing on a panel of validated sex-phenotype dependent male- and female-biased sDMRs, we tested the developmental dynamics of sex bias in liver methylation and the impacts of mutations in the androgen receptor, estrogen receptor alpha, or the transcriptional repressor Bcl6 gene. True hermaphrodites that carry both unilateral ovaries and contralateral testes were also tested. Our data show that sex bias in methylation either coincides with or follows sex bias in the expression of sDMR-proximal genes, suggesting that sex bias in gene expression may be required for demethylation at certain sDMRs. Global ablation of AR, ESR1, or a liver-specific loss of BCL6, all alter sDMR methylation, whereas presence of both an ovary and a testis delays the establishment of male-type methylation levels in hermaphrodites. Moreover, the Bcl6-LKO shows dissociation between expression and methylation, suggesting a distinct role of BCL6 in demethylation of intragenic sDMRs.


Subject(s)
DNA Methylation/genetics , Estrogen Receptor alpha/genetics , Proto-Oncogene Proteins c-bcl-6/genetics , Receptors, Androgen/genetics , Animals , Disorders of Sex Development/genetics , Epigenesis, Genetic , Female , Gene Expression Regulation/genetics , Gene Expression Regulation, Developmental/genetics , Liver/growth & development , Liver/metabolism , Male , Mice , Ovary/growth & development , Ovary/metabolism , Sex Characteristics , Sexism , Testis/growth & development , Testis/metabolism
8.
Chromosoma ; 130(1): 41-52, 2021 03.
Article in English | MEDLINE | ID: mdl-33492414

ABSTRACT

In mammalian oocytes, proper chromosome segregation at the first meiotic division is dictated by the presence and site of homologous chromosome recombination, which takes place in fetal life. Our current understanding of how homologous chromosomes find each other and initiate synapsis, which is prerequisite for homologous recombination, is limited. It is known that chromosome telomeres are anchored into the nuclear envelope (NE) at the early meiotic prophase I (MPI) and move along NE to facilitate homologous chromosome search and pairing. However, the mouse (Mus musculus) carries all acrocentric chromosomes with one telomeric end close to the centromere (subcentromeric telomere; C-telomere) and the other far away from the centromere (distal telomere; D-telomere), and how C- and D-telomeres participate in chromosome pairing and synapsis during the MPI progression is not well understood. Here, we found in the mouse oocyte that C- and D-telomeres transiently clustered in one area, but D-telomeres soon separated together from C-telomeres and then dispersed to preferentially initiate synapsis, while C-telomeres remained in clusters and synapsed at the last. In the Spo11 null oocyte, which is deficient in SPO11-dependent DSBs formation and homologous synapsis, the pattern of C- and D-telomere clustering and resolution was not affected, but synapsis was more frequently initiated at C-telomeres. These results suggest that SPO11 suppresses the early synapsis between C-telomeres in clusters.


Subject(s)
Chromosome Pairing , Chromosome Segregation , Chromosomes/genetics , Homologous Recombination , Meiotic Prophase I , Oocytes/physiology , Telomere , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Centromere , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endodeoxyribonucleases/genetics , Endodeoxyribonucleases/metabolism , Female , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Oocytes/cytology
9.
Front Genet ; 12: 792604, 2021.
Article in English | MEDLINE | ID: mdl-34987552

ABSTRACT

The sex chromosome complement, XX or XY, determines sexual differentiation of the gonadal primordium into a testis or an ovary, which in turn directs differentiation of the germ cells into sperm and oocytes, respectively, in eutherian mammals. When the X monosomy or XY sex reversal occurs, XO and XY females exhibit subfertility and infertility in the mouse on the C57BL/6J genetic background, suggesting that functional germ cell differentiation requires the proper sex chromosome complement. Using these mouse models, we asked how the sex chromosome complement affects gene transcription in the oocytes during follicular growth. An oocyte accumulates cytoplasmic components such as mRNAs and proteins during follicular growth to support subsequent meiotic progression, fertilization, and early embryonic development without de novo transcription. However, how gene transcription is regulated during oocyte growth is not well understood. Our results revealed that XY oocytes became abnormal in chromatin configuration, mitochondria distribution, and de novo transcription compared to XX or XO oocytes near the end of growth phase. Therefore, we compared transcriptomes by RNA-sequencing among the XX, XO, and XY oocytes of 50-60 µm in diameter, which were still morphologically comparable. The results showed that the X chromosome dosage limited the X-linked and autosomal gene transcript levels in XO oocytes whereas many genes were transcribed from the Y chromosome and made the transcriptome in XY oocytes closer to that in XX oocytes. We then compared the transcript levels of 3 X-linked, 3 Y-linked and 2 autosomal genes in the XX, XO, and XY oocytes during the entire growth phase as well as at the end of growth phase using quantitative RT-PCR. The results indicated that the transcript levels of most genes increased with oocyte growth while largely maintaining the X chromosome dosage dependence. Near the end of growth phase, however, transcript levels of some X-linked genes did not increase in XY oocytes as much as XX or XO oocytes, rendering their levels much lower than those in XX oocytes. Thus, XY oocytes established a distinct transcriptome at the end of growth phase, which may be associated with abnormal chromatin configuration and mitochondria distribution.

11.
Cells ; 9(6)2020 06 09.
Article in English | MEDLINE | ID: mdl-32527045

ABSTRACT

Sex biases in the genome-wide distribution of DNA methylation and gene expression levels are some of the manifestations of sexual dimorphism in mammals. To advance our understanding of the mechanisms that contribute to sex biases in DNA methylation and gene expression, we conducted whole genome bisulfite sequencing (WGBS) as well as RNA-seq on liver samples from mice with different combinations of sex phenotype and sex-chromosome complement. We compared groups of animals with different sex phenotypes, but the same genetic sexes, and vice versa, same sex phenotypes, but different sex-chromosome complements. We also compared sex-biased DNA methylation in mouse and human livers. Our data show that sex phenotype, X-chromosome dosage, and the presence of Y chromosome shape the differences in DNA methylation between males and females. We also demonstrate that sex bias in autosomal methylation is associated with sex bias in gene expression, whereas X-chromosome dosage-dependent methylation differences are not, as expected for a dosage-compensation mechanism. Furthermore, we find partial conservation between the repertoires of mouse and human genes that are associated with sex-biased methylation, an indication that gene function is likely to be an important factor in this phenomenon.


Subject(s)
DNA Methylation/genetics , Gene Expression/genetics , Liver/physiopathology , Sex Chromosomes/genetics , Animals , Female , Humans , Male , Phenotype
12.
Cell Death Dis ; 10(11): 790, 2019 10 17.
Article in English | MEDLINE | ID: mdl-31624230

ABSTRACT

Mammalian female fertility is limited by the number and quality of oocytes in the ovarian reserve. The number of oocytes is finite since all germ cells cease proliferation to become oocytes in fetal life. Moreover, 70-80% of the initial oocyte population is eliminated during fetal and neonatal development, restricting the ovarian reserve. Why so many oocytes are lost during normal development remains an enigma. In Meiotic Prophase I (MPI), oocytes go through homologous chromosome synapsis and recombination, dependent on formation and subsequent repair of DNA double strand breaks (DSBs). The oocytes that have failed in DSB repair or synapsis get eliminated mainly in neonatal ovaries. However, a large oocyte population is eliminated before birth, and the cause or mechanism of this early oocyte loss is not well understood. In the current paper, we show that the oocyte loss in fetal ovaries was prevented by a deficiency of Caspase 9 (CASP9), which is the hub of the mitochondrial apoptotic pathway. Furthermore, CASP9 and its downstream effector Caspase 3 were counteracted by endogenous X-linked Inhibitor of Apoptosis (XIAP) to regulate the oocyte population; while XIAP overexpression mimicked CASP9 deficiency, XIAP deficiency accelerated oocyte loss. In the CASP9 deficiency, more oocytes were accumulated at the pachytene stage with multiple γH2AFX foci and high LINE1 expression levels, but with normal levels of synapsis and overall DSB repair. We conclude that the oocytes with LINE1 overexpression were preferentially eliminated by CASP9-dependent apoptosis in balance with XIAP during fetal ovarian development. When such oocytes were retained, however, they get eliminated by a CASP9-independent mechanism during neonatal development. Thus, the oocyte is equipped with multiple surveillance mechanisms during MPI progression to safe-guard the quality of oocytes in the ovarian reserve.


Subject(s)
Caspase 9/metabolism , Fetal Development/physiology , Inhibitor of Apoptosis Proteins/metabolism , Oocytes/enzymology , Animals , Apoptosis/physiology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Oocytes/cytology , Ovary/cytology , Ovary/enzymology , Ovary/growth & development , Pregnancy , X-Linked Inhibitor of Apoptosis Protein/genetics , X-Linked Inhibitor of Apoptosis Protein/metabolism
13.
Am J Hum Genet ; 103(5): 740-751, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30388401

ABSTRACT

Androgenetic complete hydatidiform moles are human pregnancies with no embryos and affect 1 in every 1,400 pregnancies. They have mostly androgenetic monospermic genomes with all the chromosomes originating from a haploid sperm and no maternal chromosomes. Androgenetic complete hydatidiform moles were described in 1977, but how they occur has remained an open question. We identified bi-allelic deleterious mutations in MEI1, TOP6BL/C11orf80, and REC114, with roles in meiotic double-strand breaks formation in women with recurrent androgenetic complete hydatidiform moles. We investigated the occurrence of androgenesis in Mei1-deficient female mice and discovered that 8% of their oocytes lose all their chromosomes by extruding them with the spindles into the first polar body. We demonstrate that Mei1-/- oocytes are capable of fertilization and 5% produce androgenetic zygotes. Thus, we uncover a meiotic abnormality in mammals and a mechanism for the genesis of androgenetic zygotes that is the extrusion of all maternal chromosomes and their spindles into the first polar body.


Subject(s)
Androgens/genetics , Hydatidiform Mole/genetics , Mutation/genetics , Alleles , Animals , Chromosomes/genetics , Female , Humans , Male , Mammals/genetics , Mice , Mice, Inbred C57BL , Oocytes/pathology , Pregnancy , Zygote/pathology
15.
Sci Rep ; 7(1): 17759, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29259204

ABSTRACT

Spermatogenesis consists of a series of highly regulated processes that include mitotic proliferation, meiosis and cellular remodeling. Although alterations in gene expression are well known to modulate spermatogenesis, posttranscriptional mechanisms are less well defined. The ubiquitin proteasome system plays a significant role in protein turnover and may be involved in these posttranscriptional mechanisms. We previously identified ubiquitin ligase Huwe1 in the testis and showed that it can ubiquitinate histones. Since modulation of histones is important at many steps in spermatogenesis, we performed a complete characterization of the functions of Huwe1 in this process by examining the effects of its inactivation in the differentiating spermatogonia, spermatocytes and spermatids. Inactivation of Huwe1 in differentiating spermatogonia led to their depletion and formation of fewer pre-leptotene spermatocytes. The cell degeneration was associated with an accumulation of DNA damage response protein γH2AX, impaired downstream signalling and apoptosis. Inactivation of Huwe1 in spermatocytes indicated that Huwe1 is not essential for meiosis and spermiogenesis, but can result in accumulation of γH2AX. Collectively, these results provide a comprehensive survey of the functions of Huwe1 in spermatogenesis and reveal Huwe1's critical role as a modulator of the DNA damage response pathway in the earliest steps of spermatogonial differentiation.


Subject(s)
Cell Differentiation/physiology , Ligases/metabolism , Meiosis/physiology , Spermatogenesis/physiology , Spermatogonia/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Animals , Female , Histones/metabolism , Male , Mice , Mice, Knockout , Proteasome Endopeptidase Complex/metabolism , Spermatids/metabolism , Spermatocytes/metabolism , Spermatogonia/physiology , Testis/metabolism , Testis/physiology
16.
Sci Rep ; 7(1): 960, 2017 04 19.
Article in English | MEDLINE | ID: mdl-28424461

ABSTRACT

The B6.YTIR (XY) mouse develops bilateral ovaries despite the expression of the testis-determining gene Sry during gonadal differentiation. We reported that the oocytes of the XY female are defective in their cytoplasm, resulting in a failure in the second meiotic division after activation or fertilization in vitro. However, the mechanism of meiotic failure or the cause of infertility remained to be clarified. In the present study, we obtained mature oocytes from XY females by superovulation and confirmed that these oocytes also fail in zygotic development. By using confocal microscopy 3D-analysis, we demonstrated that meiotic spindles were properly positioned and oriented in the MII-oocytes from XY females. After parthenogenic activation, fewer oocytes from XY females extruded the second polar body, and in those oocytes, sister-chromatids were often separated but neither set entered the second polar body. ARP2, F-actin, and ORC4, known to play roles in asymmetric meiotic division, were initially localized along the ooplasmic membrane and concentrated over the MII-spindle but lost their cortical polarity after activation while the sister-chromatids moved away from the oolemma in the oocytes from XY females. Our results indicate that the second polar body extrusion is uncoupled from the sister-chromatids separation in the oocytes from XY female mouse.


Subject(s)
Chromatids/genetics , Chromosome Segregation , Cytokinesis , Gonadal Dysgenesis, 46,XY/genetics , Oocytes/physiology , Actins/metabolism , Angiopoietin-Like Protein 2 , Angiopoietin-like Proteins/metabolism , Animals , Female , Meiosis , Mice , Mice, Inbred C57BL , Oocytes/metabolism , Origin Recognition Complex/metabolism , Polar Bodies
17.
Cryobiology ; 76: 98-103, 2017 06.
Article in English | MEDLINE | ID: mdl-28412286

ABSTRACT

Oocyte cryopreservation is imperative for assisted reproductive technologies (ART). Although cryopreservation of oocytes at the Metaphase II has been widely used, immature oocytes at the germinal vesicle stage (GV-oocytes) need to be cryopreserved in certain situations such as cancer patients; however, the success rate of embryonic development from the GV-oocytes remains low largely due to the requirement for in vitro maturation (IVM). Our aim was to investigate the effects of glutathione (GSH) supplementation during vitrification and warming of mouse GV-oocytes on the preservation of developmental competence. GV-oocytes within cumulus oocyte complexes (COCs) were collected from C57BL/6J (B6) and (B6.DBA)F1 mouse strains and subjected to vitrification and warming, followed by IVM. The vitrification, warming or IVM medium was supplemented with GSH at 0-4.0 mM. In vitro matured oocytes were then fertilized in vitro and cultured in KSOMaa up to 4 days. The first cleavage and blastocyst development were evaluated morphologically, and their rates were statistically analysed by one-way ANOVA followed by Tukey's multiple comparisons test. The difference was considered significant at P < 0.05. The results showed that GSH supplementation in the IVM medium exhibited no or rather inhibitory effects on the first cleavage or blastocyst development in both mouse strains except that 1.0 mM GSH increased the blastocyst development rate in B6. By contrast, 1 mM GSH supplementation during vitrification and warming increased the blastocyst development rate in both mouse strains, more efficiently in B6 than (B6.DBA)F1. In conclusion, GSH supplementation during vitrification and warming of GV-oocytes protects the oocytes from freezing-inflicted loss of developmental competence.


Subject(s)
Cryoprotective Agents/pharmacology , Glutathione/pharmacology , Oocytes , Vitrification/drug effects , Animals , Cell Survival/drug effects , Cryopreservation/methods , Embryonic Development/drug effects , Female , Fertilization in Vitro/methods , Freezing , In Vitro Oocyte Maturation Techniques/methods , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Pregnancy
18.
PLoS One ; 10(11): e0143788, 2015.
Article in English | MEDLINE | ID: mdl-26599439

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pone.0040481.].

19.
Asian J Androl ; 17(3): 360-6, 2015.
Article in English | MEDLINE | ID: mdl-25578929

ABSTRACT

The sexual differentiation of germ cells into spermatozoa or oocytes is strictly regulated by their gonadal environment, testis or ovary, which is determined by the presence or absence of the Y chromosome, respectively. Hence, in normal mammalian development, male germ cells differentiate in the presence of X and Y chromosomes, and female germ cells do so in the presence of two X chromosomes. However, gonadal sex reversal occurs in humans as well as in other mammalian species, and the resultant XX males and XY females can lead healthy lives, except for a complete or partial loss of fertility. Germ cells carrying an abnormal set of sex chromosomes are efficiently eliminated by multilayered surveillance mechanisms in the testis, and also, though more variably, in the ovary. Studying the molecular basis for sex-specific responses to a set of sex chromosomes during gametogenesis will promote our understanding of meiotic processes contributing to the evolution of sex determining mechanisms. This review discusses the fate of germ cells carrying various sex chromosomal compositions in mouse models, the limitation of which may be overcome by recent successes in the differentiation of functional germ cells from embryonic stem cells under experimental conditions.


Subject(s)
Cell Differentiation/physiology , Germ Cells/physiology , Oocytes/physiology , Sex Chromosomes/physiology , X Chromosome/physiology , Y Chromosome/physiology , Animals , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Female , Germ Cells/cytology , Humans , Infertility, Female/physiopathology , Male , Mice , Models, Animal , Oocytes/cytology , Oogenesis/physiology , Sex Determination Processes/physiology , Spermatogenesis/physiology
20.
Hum Reprod ; 29(10): 2256-68, 2014 Oct 10.
Article in English | MEDLINE | ID: mdl-25113843

ABSTRACT

STUDY QUESTION: How does l-carnitine (LC) supplementation during vitrification and in vitro maturation (IVM) of germinal vesicle stage (GV)-oocytes improve the developmental competence of the resultant metaphase II (MII) oocytes? SUMMARY ANSWER: LC supplementation during both vitrification of GV-oocytes and their subsequent IVM improved nuclear maturation as well as meiotic spindle assembly and mitochondrial distribution in MII oocytes. WHAT IS KNOWN ALREADY: Vitrification of GV-oocytes results in a lower success rate of blastocyst development compared with non-vitrified oocytes. LC supplementation during both vitrification and IVM of mouse GV-oocytes significantly improves embryonic development after IVF. STUDY DESIGN, SIZE, DURATION: GV-oocytes were collected from (B6.DBA)F1 and B6 mouse strains and subjected to vitrification and warming with or without 3.72 mM LC supplementation. After IVM with or without LC supplementation, the rate of nuclear maturation and the quality of MII oocytes were evaluated. At least 20 oocytes/group were examined, and each experiment was repeated at least three times. All experiments were conducted during 2013-2014. PARTICIPANTS/MATERIALS, SETTING, METHODS: Extrusion of the first polar body in IVM oocytes was observed as an indication of nuclear maturation. Spindle assembly and chromosomal alignment were examined by immunostaining of α-tubulin and nuclear staining with 4,6-diamidino-2-phenylindole (DAPI). Mitochondrial distribution and oxidative activity were measured by staining with Mitotracker Green Fluorescence Mitochondria (Mitotracker Green FM) and chloromethyltetramethylrosamine (Mitotracker Orange CMTMRos), respectively. ATP levels were determined by using the Bioluminescent Somatic Cell Assay Kit. MAIN RESULTS AND THE ROLE OF CHANCE: LC supplementation during both vitrification and IVM of GV-oocytes significantly increased the proportions of oocytes with normal MII spindles to the levels comparable with those of non-vitrified oocytes in both mouse strains. While vitrification of GV-oocytes lowered the proportions of MII oocytes with peripherally concentrated mitochondrial distribution compared with non-vitrified oocytes, LC supplementation significantly increased the proportion of such oocytes in the (B6.DBA)F1 strain. LC supplementation decreased the proportion of oocytes with mitochondrial aggregates in both vitrified and non-vitrified oocytes in the B6 strain. The oxidative activity of mitochondria was mildly decreased by vitrification and drastically increased by LC supplementation irrespective of vitrification in both mouse strains. No change was found in ATP levels irrespective of vitrification or LC supplementation. Results were considered to be statistically significant at P < 0.05 by either χ(2)- or t-test. LIMITATIONS, REASONS FOR CAUTION: It remains to be tested whether beneficial effect of LC supplementation during vitrification and IVM of GV-oocytes leads to fetal development and birth of healthy offspring after embryo transfer to surrogate females. WIDER IMPLICATIONS OF THE FINDINGS: This protocol has the potential to improve the quality of vitrified human oocytes and embryos during assisted reproduction treatment. STUDY FUNDING/COMPETING INTEREST: Partially supported by the Natural Sciences and Engineering Research Council of Canada (NSERC) Discovery Grant and Mitacs Elevate Postdoctoral Fellowship, Canada.


Subject(s)
Carnitine/pharmacology , In Vitro Oocyte Maturation Techniques , Metaphase/drug effects , Mitochondria/drug effects , Spindle Apparatus/drug effects , Vitrification , Adenosine Triphosphate/metabolism , Animals , Cell Culture Techniques , Female , Male , Mice , Mice, Inbred DBA , Mitochondria/ultrastructure , Oocytes/growth & development , Spindle Apparatus/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...