Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Placenta ; 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38944560

ABSTRACT

Large, multinucleated cells, like syncytiotrophoblasts (STB), are not readily analyzed by standard methods used for single cells, such as single-cell RNA-sequencing and fluorescence-activated cellular sorting (FACS). Here we have demonstrated that fluorescence-activated nuclear sorting (FANS) is suitable to analyze nuclei from STB. Human pluripotent stem cells (PSCs) can be differentiated into a mixed trophoblast populations comprising approximately 20 % STB by treatment with BMP4 (Bone Morphogenetic Protein-4), plus A83-01 and PD173074, inhibitors of activin and FGF2 signaling, respectively (the BAP model) in about a week. Here we demonstrate that FANS can be used to separate two types of STB nuclei from the nine different clusters of trophoblast nuclei previously identified in the BAP model by single nucleus RNA sequencing (snRNAseq). Rather than using cell surface markers, as in FACS, transcription factors in various combinations were employed to target specific nuclear types. Nuclei were isolated at d 8 of BAP differentiation of H1 human embryonic stem cells and fixed in 4 % paraformaldehyde. After permeabilization in 0.1 % triton X-100, nuclei were incubated for 3 and 1 h at 4 °C with primary and secondary antibodies respectively and nuclear samples were then subjected to FANS. By using markers identified by snRNA and immunohistochemistry, nuclei were first sorted into a Topoisomerase-1, or TOP1, bright population and then into the two STB subpopulations by using antibodies to JUNB (Jun B Proto-Oncogene) and TFCP2L1 (Transcription Factor CP2 Like 1). The protocol established here is simple, straightforward, and efficient and can be used on a relatively large scale to sort individual subtypes of nuclei from mixed populations of trophoblasts for further analysis.

2.
Stem Cell Reports ; 17(6): 1289-1302, 2022 06 14.
Article in English | MEDLINE | ID: mdl-35594861

ABSTRACT

The observation that trophoblast (TB) can be generated from primed pluripotent stem cells (PSCs) by exposure to bone morphogenetic protein-4 (BMP4) when FGF2 and ACTIVIN signaling is minimized has recently been challenged with the suggestion that the procedure instead produces amnion. Here, by analyzing transcriptome data from multiple sources, including bulk and single-cell data, we show that the BMP4 procedure generates bona fide TB with similarities to both placental villous TB and TB generated from TB stem cells. The analyses also suggest that the transcriptomic signatures between embryonic amnion and different forms of TB have commonalities. Our data provide justification for the continued use of TB derived from PSCs as a model for investigating placental development.


Subject(s)
Pluripotent Stem Cells , Trophoblasts , Amnion , Cell Differentiation , Embryonic Stem Cells , Female , Humans , Placenta , Pregnancy
3.
Front Genet ; 13: 872818, 2022.
Article in English | MEDLINE | ID: mdl-35432469

ABSTRACT

Understanding the process of human placentation is important to the development of strategies for treatment of pregnancy complications. Several animal and in vitro human model systems for the general study human placentation have been used. The field has expanded rapidly over the past decades to include stem cell-derived approaches that mimic preclinical placental development, and these stem cell-based models have allowed us to better address the physiology and pathophysiology of normal and compromised trophoblast (TB) sublineage development. The application of transcriptomic approaches to these models has uncovered limitations that arise when studying the distinctive characteristics of the large and fragile multinucleated syncytiotrophoblast (STB), which plays a key role in fetal-maternal communication during pregnancy. The extension of these technologies to induced pluripotent stem cells (iPSCs) is just now being reported and will allow, for the first time, a reproducible and robust approach to the study of the developmental underpinnings of late-manifesting diseases such as preeclampsia (PE) and intrauterine growth retardation in a manner that is patient- and disease-specific. Here, we will first focus on the application of various RNA-seq technologies to TB, prior limitations in fully accessing the STB transcriptome, and recent leveraging of single nuclei RNA sequencing (snRNA-seq) technology to improve our understanding of the STB transcriptome. Next, we will discuss new stem-cell derived models that allow for disease- and patient-specific study of pregnancy disorders, with a focus on the study of STB developmental abnormalities in PE that combine snRNA-seq approaches and these new in vitro models.

4.
Reprod Biomed Online ; 43(5): 913-919, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34493464

ABSTRACT

RESEARCH QUESTION: Male infertility is a global issue worldwide and multiple morphological abnormalities of the sperm flagella (MMAF) is one of the most severe forms of the qualitative sperm defects with a heterogeneous genetic cause that has not been completely understood. Can whole-exome sequencing (WES) reveal novel genetic causes contributing to MMAF in a consanguineous Pakistani family, comprising three infertile brothers? DESIGN: WES and bioinformatic analysis were conducted to screen potential pathogenic variants. The identified variant was validated by Sanger sequencing in all available family members Transmission electron microscopy analyses was carried out to examine the flagella ultrastructure of spermatozoa from patient. RESULTS: WES and Sanger sequencing identified a novel homozygous stop-gain mutation (ENST00000392644.4, c.182C>G, p.S61X) in ARMC2, which is expected to lead to loss of protein functions. Transmission electron microscopy analyses revealed that the flagellar ultrastructure of the patient's spermatozoa was disorganized along with a complete absence of central pair complex (CPC), suggesting that ARMC2 is involved in the assembly, stability of the axonemal complex, or both, particularly the CPC. CONCLUSION: We report that a familial stop-gain mutation in ARMC2 is associated with male infertility in humans caused by MMAF accompanied with loss of CPCs and axonemal disorganization. We provide genetic evidence that ARMC2 is essential for human spermatogenesis and its mutation may be pathogenic for MMAF. These findings will improve the knowledge about the genetic basis of MMAF and provide information for genetic counselling of this disease.


Subject(s)
Cytoskeletal Proteins/genetics , Sperm Tail/pathology , Spermatozoa/abnormalities , Adult , Consanguinity , Cytoskeletal Proteins/physiology , Homozygote , Humans , Infertility, Male/genetics , Male , Microscopy, Electron, Transmission , Mutation , Pakistan , Pedigree , Semen Analysis , Spermatogenesis , Spermatozoa/ultrastructure , Exome Sequencing
5.
Front Cell Dev Biol ; 9: 695248, 2021.
Article in English | MEDLINE | ID: mdl-34368143

ABSTRACT

One model to study the emergence of the human trophoblast (TB) has been the exposure of pluripotent stem cells to bone morphogenetic protein 4 (BMP4) in presence of inhibitors of ACTIVIN/TGFB; A83-01 and FGF2; PD173074 (BAP), which generates a mixture of cytotrophoblast, syncytiotrophoblast, and cells with similarities to extravillous trophoblast. Here, H1 human embryonic stem cells were BAP-exposed under two O2 conditions (20% and 5%, respectively). At day 8, single nuclei RNA sequencing was used for transcriptomics analysis, thereby allowing profiling of fragile syncytial structures as well as the more resilient mononucleated cells. Following cluster analysis, two major groupings, one comprised of five (2,4,6,7,8) and the second of three (1,3,5) clusters were evident, all of which displayed recognized TB markers. Of these, two (2 and 3) weakly resembled extravillous trophoblast, two (5 and 6) strongly carried the hallmark transcripts of syncytiotrophoblast, while the remaining five were likely different kinds of mononucleated cytotrophoblast. We suggest that the two populations of nuclei within syncytiotrophoblast may have arisen from fusion events involving two distinct species of precursor cells. The number of differentially expressed genes between O2 conditions varied among the clusters, and the number of genes upregulated in cells cultured under 5% O2 was highest in syncytiotrophoblast cluster 6. In summary, the BAP model reveals an unexpectedly complex picture of trophoblast lineage emergence that will need to be resolved further in time-course studies.

6.
Placenta ; 113: 8-14, 2021 09 15.
Article in English | MEDLINE | ID: mdl-33504453

ABSTRACT

Three versions of syncytiotrophoblast exist in the human placenta: an invasive type associated with the implanting conceptus, non-invasive villous type of definitive placenta, and placental bed giant cells. Syncytins are encoded by modified env genes of endogenous retroviruses (ERV), but how they contribute functionally to placental syncytial structures is unclear. A minimum of eight genes (ERVW1, ERVFRD-1, ERVV-1, ERVV-2, ERVH48-1, ERVMER34-1, ERV3-1, & ERVK13-1) encoding syncytin family members are expressed in human trophoblast, the majority from implantation to term. ERVW1 (Syncytin 1) and ERVFRD-1 (Syncytin 2) are considered the major fusogens, but, when the expression of their genes is analyzed by single cell RNAseq in first trimester placenta, their transcripts are distinctly patterned and also differ from those of their proposed binding partners, SLC1A5 and MFSD2A, respectively. ERVRH48-1 (suppressyn or SUPYN) and ERVMER34-1 are probable negative regulators of fusion and co-expressed, primarily in cytotrophoblast. The remaining genes and their products have been little studied. Syncytin expression is a feature of placental development in almost all eutherian mammals studied, in at least one marsupial, and in viviparous lizards, which lack the trophoblast lineage. Their expression has been inferred to be essential for pregnancy success in the mouse. All the main human ERV genes arose following independent retroviral insertion events, none of which trace back to the divergence of eutherians and metatherians (marsupials). While syncytins may be crucial for placental development, it seems unlikely that they helped orchestrate the divergence of eutherians and marsupials.


Subject(s)
Biological Evolution , Endogenous Retroviruses/genetics , Gene Products, env/metabolism , Placentation , Pregnancy Proteins/metabolism , Trophoblasts/metabolism , Cell Fusion , Female , Gene Products, env/genetics , Humans , Pregnancy , Pregnancy Proteins/genetics
7.
J Exp Med ; 217(2)2020 02 03.
Article in English | MEDLINE | ID: mdl-31658987

ABSTRACT

Asthenozoospermia is a common cause of male infertility, but its etiology remains incompletely understood. We recruited three Pakistani infertile brothers, born to first-cousin parents, displaying idiopathic asthenozoospermia but no ciliary-related symptoms. Whole-exome sequencing identified a missense variant (c.G5408A, p.C1803Y) in DNAH17, a functionally uncharacterized gene, recessively cosegregating with asthenozoospermia in the family. DNAH17, specifically expressed in testes, was localized to sperm flagella, and the mutation did not alter its localization. However, spermatozoa of all three patients showed higher frequencies of microtubule doublet(s) 4-7 missing at principal piece and end piece than in controls. Mice carrying a homozygous mutation (Dnah17M/M) equivalent to that in patients recapitulated the defects in patients' sperm tails. Further examinations revealed that the doublets 4-7 were destabilized largely due to the storage of sperm in epididymis. Altogether, we first report that a homozygous DNAH17 missense variant specifically induces doublets 4-7 destabilization and consequently causes asthenozoospermia, providing a novel marker for genetic counseling and diagnosis of male infertility.


Subject(s)
Asthenozoospermia/genetics , Axonemal Dyneins/genetics , Mutation, Missense , Sperm Tail/pathology , Adult , Animals , Asthenozoospermia/pathology , Cell Line, Tumor , Disease Models, Animal , Female , Genotype , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Spermatozoa/pathology , Testis/pathology , Transfection
8.
Horm Res Paediatr ; 91(1): 9-16, 2019.
Article in English | MEDLINE | ID: mdl-30947225

ABSTRACT

BACKGROUND: Congenital hypogonadotropic hypogonadism (CHH) is a heterogeneous disorder characterized by delayed or loss of puberty and infertility due to functional deficiency in the hypothalamic gonadotropin-releasing hormone (GnRH). CHH can be classified into 2 subtypes on the basis of olfaction: Kallmann syndrome and normosmic CHH (nCHH). The spectrum of genetic variants causing CHH is continually expanding. Here, we recruited a consanguineous Pakistani family having 2 male and 2 female infertile patients diagnosed with idiopathic nCHH. AIMS: The aim of this study was to investigate the genetic cause of nCHH in the family. METHODS: Clinical and physical analyses were performed for the patients. Genetic analysis was carried out using whole exome and Sanger sequencing. RESULTS: Clinical and physical investigations confirmed low levels of gonadotropins and failure of secondary sexual development in the patients. Genetic analysis identified a novel nonsense mutation (chr4: g.68619942G>A, c.112C>T, p.Arg38*) in the gonadotropin-releasing hormone receptor gene (GNRHR) recessively co-segregating with nCHH in this family. All the patients are homozygous and their parents are heterozygous carriers, while normal siblings are heterozygous carriers or wild-type for this mutation, indicating that the identified mutation is pathogenic for nCHH in the family. CONCLUSION: We report the first homozygous nonsense mutation in the GNRHR gene (chr4: g. 68619942G>A, c.112C>T, p. Arg38*) that is associated with familial nCHH. Hence, our study displayed a good correlation of the genotype and phenotype of nCHH patients.


Subject(s)
Codon, Nonsense , Exome , Family , Infertility, Female/genetics , Infertility, Male/genetics , Kallmann Syndrome/genetics , Receptors, LHRH/genetics , Adult , Female , Humans , Male , Pakistan , Exome Sequencing
9.
Sci China Life Sci ; 62(4): 544-552, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30919279

ABSTRACT

The balanced actions between ubiquitination and deubiquitination precisely control the levels of various proteins vital for spermatogenesis. Ubiquitin-specific processing proteases (USPs) are the largest family of deubiquitinatingenzymes(DUBs), containing more than 50 members. So far, the functions of only a few USPs in male fertility have been studied, the roles of the majority are yet unknown. The present study aimed to explore the function of Usp29 (ubiquitin-specific protease 29) in male fertility. We found that Usp29 showed predominant expression in mouse testis, and its mRNA expression started to increase at 14 days postpartum (dpp), with a peak at 28 and 35 dpp. Using CRISPR/Cas9 technology, we generated Usp29 knockout mice (Usp29-/-). Usp29-/- mice exhibited no overt developmental anomalies. Further examination revealed that Usp29-/- mice had normal fertility and showed no detectable difference in the testis/body weight ratio, testicular and epididymal histology as well as epididymal sperm count from the wild-type littermates. Moreover, Usp29 is not a pseudogene in mice. Taken together, our study first reported that though Usp29 is predominantly expressed in the testis, it is not essential for male fertility in mice.


Subject(s)
Fertility/genetics , Ubiquitin-Specific Proteases/metabolism , Animals , Epididymis/anatomy & histology , Female , Genome/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phylogeny , RNA, Messenger/metabolism , Sperm Count , Spermatogenesis , Testis/anatomy & histology , Testis/metabolism , Ubiquitin-Specific Proteases/deficiency , Ubiquitin-Specific Proteases/genetics
10.
Genet Med ; 21(1): 62-70, 2019 01.
Article in English | MEDLINE | ID: mdl-29895858

ABSTRACT

PURPOSE: Fanconi anemia (FA) genes play important roles in spermatogenesis. In mice, disruption of Fancm impairs male fertility and testicular integrity, but whether FANCM pathogenic variants (PV) similarly affect fertility in men is unknown. Here we characterize a Pakistani family having three infertile brothers, two manifesting oligoasthenospermia and one exhibiting azoospermia, born to first-cousin parents. A homozygous PV in FANCM (c.1946_1958del, p.P648Lfs*16) was found cosegregating with male infertility. Our objective is to validate that FANCM p.P648Lfs*16 is the PV causing infertility in this family. METHODS: Exome and Sanger sequencing were used for PV screening. DNA interstrand crosslink (ICL) sensitivity was assessed in lymphocytes from patients. A mouse model carrying a PV nearly equivalent to that in the patients (FancmΔC/ΔC) was generated, followed by functional analysis in spermatogenesis. RESULTS: The loss-of-function FANCM PV increased ICL sensitivity in lymphocytes of patients and FancmΔC/ΔC spermatogonia. Adult FancmΔC/ΔC mice showed spermatogenic failure, with germ cell loss in 50.2% of testicular tubules and round-spermatid maturation arrest in 43.5% of tubules. In addition, neither bone marrow failure nor cancer/tumor was detected in all the patients or adult FancmΔC/ΔC mice. CONCLUSION: These findings revealed male infertility to be a novel phenotype of human patients with a biallelic FANCM PV.


Subject(s)
DNA Helicases/genetics , Genetic Predisposition to Disease , Infertility, Male/genetics , Spermatogenesis/genetics , Adult , Animals , Frameshift Mutation , Homozygote , Humans , Infertility, Male/pathology , Loss of Function Mutation/genetics , Male , Mice , Oligospermia/genetics , Oligospermia/pathology , Pedigree , Phenotype , Testis/pathology
11.
Genet Med ; 21(1): 266, 2019 01.
Article in English | MEDLINE | ID: mdl-30158692

ABSTRACT

Hao Win, Hui Ma and Sajjad Hussain were incorrectly affiliated to 'Department of Radiation Oncology, The Houston Methodist Research Institute, Houston, TX 77030 USA'. These authors should only have been affiliated to 'Hefei National Laboratory for Physical Sciences at Microscale, The First Affiliated Hospital of USTC, USTC-SJH Joint Center for Human Reproduction and Genetics, The CAS Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei 230027, China'. They were also not noted as contributing equally to the paper. Both these errors have now been corrected in the PDF and HTML versions of the paper.

12.
J Hum Genet ; 63(10): 1071-1076, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30033443

ABSTRACT

Hereditary leukonychia (also known as porcelain nails or white nails) is a genetic disorder. It may exist as an isolated feature or associated with other cutaneous or systemic disorders. Although a number of genes have been described to cause leukonychia, still the underlying genetic etiologies of many cases remain unknown. Here, we report a Pakistani family presenting leukonychia and koilonychia nails in mother and five of her kids. All the affected individuals had white to pale nails in appearance exhibiting complete and partial leukonychia, respectively. Similarly, nails of finger and toe appeared brittle and concave, showing the characteristics features of koilonychia. Whole exome sequencing and subsequent Sanger sequencing identified a pathogenic novel missense mutation (c.1390G>A, p.Glu464Lys) in PLCD1, co-segregating with the disorder in an autosomal dominant pattern. In silico prediction tools supported the pathogenicity of the identified mutation. Literature review determined that mutations in PLCD1 only cause leukonychia. Therefore, our findings add another pathogenic variant to the PLCD1 mutation pool causing leukonychia that would help to understand the underlying molecular mechanism.


Subject(s)
Exome Sequencing , Family , Genes, Dominant , Hypopigmentation/genetics , Mutation, Missense , Nail Diseases/congenital , Phospholipase C delta/genetics , Female , Humans , Hypopigmentation/pathology , Male , Nail Diseases/genetics , Nail Diseases/pathology
13.
PLoS Genet ; 14(5): e1007300, 2018 05.
Article in English | MEDLINE | ID: mdl-29795555

ABSTRACT

Three waves of H2AX phosphorylation (γH2AX) have been observed in male meiotic prophase I: the first is ATM-dependent and occurs at leptonema, while the second and third are ATR-dependent, occuring at zygonema and pachynema, respectively. The third wave of H2AX phosphorylation marks and silences unsynapsed chromosomes. Little is known about H2AX phosphorylation expands to chromatin-wide regions in spermatocytes. Here, we report that histone acetyltransferase (HAT) MOF is involved in all three waves of H2AX phosphorylation expansion. Germ cell-specific deletion of Mof in spermatocytes by Stra8-Cre (Mof cKO) caused global loss of H4K16ac. In leptotene and zygotene spermatocytes of cKO mice, the γH2AX signals were observed only along the chromosomal axes, and chromatin-wide H2AX phosphorylation was lost. In almost 40% of early-mid pachytene spermatocytes from Mof cKO mice, γH2AX and MDC1 were detected along the unsynapsed axes of the sex chromosomes, but failed to expand, which consequently caused meiotic sex chromosome inactivation (MSCI) failure. Furthermore, though RAD51 was proficiently recruited to double-strand break (DSB) sites, defects in DSB repair and crossover formation were observed in Mof cKO spermatocytes, indicating that MOF facilitates meiotic DSB repair after RAD51 recruitment. We propose that MOF regulates male meiosis and is involved in the expansion of all three waves of H2AX phosphorylation from the leptotene to pachytene stages, initiated by ATM and ATR, respectively.


Subject(s)
Histone Acetyltransferases/metabolism , Histones/metabolism , Spermatogenesis/physiology , Animals , DNA Breaks, Double-Stranded , DNA Repair , Female , Histone Acetyltransferases/genetics , Male , Meiosis , Mice, Knockout , Mice, Transgenic , Pachytene Stage , Phosphorylation , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Spermatocytes/cytology , Testis/physiology
14.
Sci Rep ; 8(1): 4975, 2018 03 21.
Article in English | MEDLINE | ID: mdl-29563520

ABSTRACT

There are more than 2300 genes that are predominantly expressed in mouse testes. The role of hundreds of these genes has been studied in mouse spermatogenesis but still there are many genes whose function is unknown. Gene knockout (KO) strategy in mice is widely used for in vivo study of gene function. The present study was designed to explore the function of the four genes: Tex37, Ccdc73, Prss55 and Nxt2, which were evolutionarily conserved in eutherians. We found that these genes had a testis-enriched expression pattern in mice except Nxt2. We knocked out these genes by CRISPR/Cas9 individually and found that all the KO mice had normal fertility with no detectable difference in testis/body weight ratios, epididymal sperm counts, as well as testicular and epididymal histology from wild type mice. Although these genes are evolutionarily conserved in eutherians including human and mouse, they are not individually essential for spermatogenesis, testis development and male fertility in mice in laboratory conditions. Our report of these fertile KO data could avoid the repetition and duplication of efforts which will help in prioritizing efforts to focus on genes that are indispensable for male reproduction.


Subject(s)
Conserved Sequence/physiology , Fertility/physiology , Proteins/physiology , Serine Proteases/physiology , Spermatogenesis/physiology , Animals , CRISPR-Cas Systems/genetics , Conserved Sequence/genetics , Epididymis/physiology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Proteins/genetics , Serine Proteases/genetics , Sperm Count , Testis/physiology
15.
Curr Biol ; 27(10): 1498-1505.e6, 2017 May 22.
Article in English | MEDLINE | ID: mdl-28502657

ABSTRACT

The mammalian sex chromosomes have undergone profound changes during their evolution from an ancestral pair of autosomes [1-4]. Specifically, the X chromosome has acquired a paradoxical sex-biased function by redistributing gene contents [5, 6] and has generated a disproportionately high number of retrogenes that are located on autosomes and exhibit male-biased expression patterns [6]. Several selection-based models have been proposed to explain this phenomenon, including a model of sexual antagonism driving X inactivation (SAXI) [6-8] and a compensatory mechanism based on meiotic sex chromosome inactivation (MSCI) [6, 8-11]. However, experimental evidence correlating the function of X-chromosome-derived autosomal retrogenes with evolutionary forces remains limited [12-17]. Here, we show that the deficiency of Rpl10l, a murine autosomal retrogene of Rpl10 with testis-specific expression, disturbs ribosome biogenesis in late-prophase spermatocytes and prohibits the transition from prophase into metaphase of the first meiotic division, resulting in male infertility. Rpl10l expression compensates for the lack of Rpl10, which exhibits a broad expression pattern but is subject to MSCI during spermatogenesis. Importantly, ectopic expression of RPL10L prevents the death of cultured RPL10-deficient somatic cells, and Rpl10l-promoter-driven transgenic expression of Rpl10 in spermatocytes restores spermatogenesis and fertility in Rpl10l-deficient mice. Our results demonstrate that Rpl10l plays an essential role during the meiotic stage of spermatogenesis by compensating for MSCI-mediated transcriptional silencing of Rpl10. These data provide direct evidence for the compensatory hypothesis and add novel insight into the evolution of X-chromosome-derived autosomal retrogenes and their role in male fertility.


Subject(s)
Meiosis , Ribosomal Proteins/metabolism , Spermatogenesis , X Chromosome Inactivation , Animals , Cell Proliferation , Cells, Cultured , Female , HEK293 Cells , Humans , Infertility, Male/metabolism , Infertility, Male/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Inbred ICR , Mice, Knockout , Mice, Transgenic , Phylogeny , Ribosomal Protein L10 , Ribosomes/metabolism , Spermatocytes/cytology , Spermatocytes/physiology , Testis/cytology , Testis/physiology
16.
Development ; 144(12): 2165-2174, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28506985

ABSTRACT

Proper oocyte development is crucial for female fertility and requires timely and accurate control of gene expression. K (lysine) acetyltransferase 8 (KAT8), an important component of the X chromosome dosage compensation system in Drosophila, regulates gene activity by acetylating histone H4 preferentially at lysine 16. To explore the function of KAT8 during mouse oocyte development, we crossed Kat8flox/flox mice with Gdf9-Cre mice to specifically delete Kat8 in oocytes. Oocyte Kat8 deletion resulted in female infertility, with follicle development failure in the secondary and preantral follicle stages. RNA-seq analysis revealed that Kat8 deficiency in oocytes results in significant downregulation of antioxidant genes, with a consequent increase in reactive oxygen species. Intraperitoneal injection of the antioxidant N-acetylcysteine rescued defective follicle and oocyte development resulting from Kat8 deficiency. Chromatin immunoprecipitation assays indicated that KAT8 regulates antioxidant gene expression by direct binding to promoter regions. Taken together, our findings demonstrate that KAT8 is essential for female fertility by regulating antioxidant gene expression and identify KAT8 as the first histone acetyltransferase with an essential function in oogenesis.


Subject(s)
Histone Acetyltransferases/metabolism , Oogenesis/physiology , Reactive Oxygen Species/metabolism , Animals , Antioxidants/metabolism , Apoptosis , Female , Fertility/genetics , Fertility/physiology , Gene Expression Regulation, Developmental , Granulosa Cells/metabolism , Heterochromatin/genetics , Heterochromatin/metabolism , Histone Acetyltransferases/deficiency , Histone Acetyltransferases/genetics , Infertility, Female/genetics , Infertility, Female/metabolism , Infertility, Female/pathology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Oocytes/cytology , Oocytes/metabolism , Oogenesis/genetics , Ovarian Follicle/cytology , Ovarian Follicle/metabolism , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL
...