Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
Add more filters










Publication year range
1.
Development ; 151(8)2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38691389

ABSTRACT

Mammalian spermatogenesis, probably the most complex of all cellular developmental processes, is an ideal model both for studying the specific mechanism of gametogenesis and for understanding the basic rules governing all developmental processes, as it entails both cell type-specific and housekeeping molecular processes. Spermatogenesis can be viewed as a mission with many tasks to accomplish, and its success is genetically programmed and ensured by the collaboration of a large number of genes. Here, I present an overview of mammalian spermatogenesis and the mechanisms underlying each step in the process, covering the cellular and molecular activities that occur at each developmental stage and emphasizing their gene regulation in light of recent studies.


Subject(s)
Gene Expression Regulation, Developmental , Spermatogenesis , Animals , Humans , Male , Mammals/genetics
2.
Cell Prolif ; 57(5): e13580, 2024 May.
Article in English | MEDLINE | ID: mdl-38230761

ABSTRACT

The mammalian Pre-B cell leukaemia transcription factors 1-4 (PBX1-4) constitutes the PBC class of the homeodomain (HD)-containing proteins, which play important roles in diverse developmental processes. The functions and the underlying molecular mechanisms of PBX1-3 but not PBX4 have been extensively studied, and they have been reported to direct essential morphogenetic processes and organogenesis. In the present study, we generated knockin mice of FLAG-tagged PBX4 and the Pbx4 knockout (KO) mice and carried out in-depth characterisation of PBX4 expression and function. PBX4 was initially detected only in the testis among several organs of the adult mice and was expressed in spermatocytes and spermatids. However, no abnormality in spermatogenesis, but growth retardation and premature death after birth were observed in most adult Pbx4 KO mice. These animals were inactive and had shorter hindlimbs and lower numbers of reticulocytes and lymphocytes, probably caused by abnormalities at earlier developmental stages. Pbx4 mRNAs were indeed detected in several embryonic cell types related to limb development by in situ hybridisation and single-cell RNA-sequencing analysis. Pbx4 protein was also detected in the bone marrow of adult mice with a lower level compared with that in the testis. PBX4 preferentially binds to the promoters of a large number of genes including those for other HD-containing proteins and ribosomal proteins whose mutations are related to anaemia. PBX4-binding sites are enriched in motifs similar to those of other HD-containing proteins such as PKNOX1 indicating that PBX4 may also act as a co-transcription factor like other PBC proteins. Together, these results show that PBX4 participates in limb development and haematopoiesis while its function in spermatogenesis has not been revealed by gene KO probably due to the complementary effects of other genes.


Subject(s)
DNA-Binding Proteins , Extremities , Gene Expression Regulation, Developmental , Hematopoiesis , Homeodomain Proteins , Animals , Male , Mice , Hematopoiesis/genetics , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic/genetics , Testis/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism
3.
Sheng Wu Gong Cheng Xue Bao ; 39(10): 4108-4122, 2023 Oct 25.
Article in Chinese | MEDLINE | ID: mdl-37877394

ABSTRACT

Meiotic initiation is a critical step in gametogenesis. Recently, some genes required for meiotic initiation have been identified. However, meiosis-initiating factors and the underlying mechanisms are far from being fully understood. We have established a long-term culture system of spermatogonial stem cells (SSCs) and an in vitro model of meiotic initiation using mouse SSCs. Our previous study revealed that the RNA-binding protein RBFOX2 may regulate meiotic initiation, but the role and the mechanism need to be further elucidated. In this study, we constructed RBFOX2 knockdown SSC lines by using lentivirus-mediated gene delivery method, and found that the knockdown SSCs underwent normal self-renewal, mitosis and differentiation. However, they were unable to initiate meiosis when treated with retinoic acid, and they underwent apoptosis. These results indicate that RBFOX2 plays an essential role in meiotic initiation of spermatogonia. This work provides new clues for understanding the functions of RNA-binding proteins in meiotic initiation.


Subject(s)
Meiosis , Spermatogonia , Mice , Male , Animals , Spermatogonia/metabolism , Meiosis/genetics , Cell Differentiation , Tretinoin/metabolism , Tretinoin/pharmacology , Mitosis , Testis/metabolism
4.
Cell Death Differ ; 30(9): 2187-2199, 2023 09.
Article in English | MEDLINE | ID: mdl-37543710

ABSTRACT

ARID1A, an SWI/SNF chromatin-remodeling gene, is commonly mutated in cancer and hypothesized to be a tumor suppressor. Recently, loss-of-function of ARID1A gene has been shown to cause intellectual disability. Here we generate Arid1a conditional knockout mice and investigate Arid1a function in the hippocampus. Disruption of Arid1a in mouse forebrain significantly decreases neural stem/progenitor cells (NSPCs) proliferation and differentiation to neurons within the dentate gyrus (DG), increasing perinatal and postnatal apoptosis, leading to reduced hippocampus size. Moreover, we perform single-cell RNA sequencing (scRNA-seq) to investigate cellular heterogeneity and reveal that Arid1a is necessary for the maintenance of the DG progenitor pool and survival of post-mitotic neurons. Transcriptome and ChIP-seq analysis data demonstrate that ARID1A specifically regulates Prox1 by altering the levels of histone modifications. Overexpression of downstream target Prox1 can rescue proliferation and differentiation defects of NSPCs caused by Arid1a deletion. Overall, our results demonstrate a critical role for Arid1a in the development of the hippocampus and may also provide insight into the genetic basis of intellectual disabilities such as Coffin-Siris syndrome, which is caused by germ-line mutations or microduplication of Arid1a.


Subject(s)
Abnormalities, Multiple , Neoplasms , Animals , Female , Mice , Pregnancy , Abnormalities, Multiple/genetics , Chromatin , Chromatin Assembly and Disassembly , Dentate Gyrus , Nuclear Proteins/metabolism
5.
Front Cell Dev Biol ; 11: 1154938, 2023.
Article in English | MEDLINE | ID: mdl-37215089

ABSTRACT

MicroRNAs (miRNAs) are believed to play important roles in mammalian spermatogenesis mainly because spermatogenesis is more or less disrupted when genes encoding key enzymes for miRNA biogenesis are mutated. However, it is challenging to study the functions of individual miRNAs due to their family-wise high sequence similarities and the clustered genomic distributions of their genes, both of which expose difficulties in using genetic methods. Accumulating evidence shows that a number of miRNAs indeed play important roles in mammalian spermatogenesis and the underlying mechanisms start to be understood. In this mini review, we focus on highlighting the roles of miRNAs in mammalian spermatogenesis elucidated mainly by using in vivo genetic methods and on discussing the underlying mechanisms. We propose that studies on the roles of miRNAs in spermatogenesis should and can be conducted in a more fruitful way given the progress in traditional methods and the birth of new technologies.

7.
FASEB J ; 36(10): e22522, 2022 10.
Article in English | MEDLINE | ID: mdl-36052752

ABSTRACT

Spermatogenesis is a highly coordinated process that initiates shortly after birth and continues throughout the lifespan of male animals. Foxo1 is a transcription factor and is involved in many biological processes. It has been reported that the inactivation of Foxo1 in gonocytes during the embryonic stage causes the defects of spermatogenesis. In the present study, we found that the inactivation of Foxo1 in spermatogonia after birth also caused germ cell loss and male infertility. We found that the initiation of meiosis was not affected; however, the germ cell development was arrested after meiosis and lack of mature spermatozoa in the cauda epididymis. We also found that the proliferation of Foxo1-deficient spermatogonia stem cells was significantly reduced under in vitro conditions. Further study revealed that inactivation of Pten in postnatal spermatogonia using Stra8-Cre did not affect germ cell development and the subcellular location of FOXO1 in Pten-deficient spermatogonia. This study demonstrated that Foxo1 was involved in the development of spermatogonia after birth and the function of Foxo1 was probably not regulated by PI3K/PTEN signaling.


Subject(s)
Phosphatidylinositol 3-Kinases , Spermatogonia , Animals , Male , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/physiology , Spermatogenesis/physiology , Spermatogonia/metabolism , Testis/metabolism
8.
EMBO Rep ; 23(8): e54298, 2022 08 03.
Article in English | MEDLINE | ID: mdl-35712867

ABSTRACT

MicroRNAs (miRNAs) are believed to play important roles in mammalian spermatogenesis but the in vivo functions of single miRNAs in this highly complex developmental process remain unclear. Here, we report that miR-202, a member of the let-7 family, plays an important role in spermatogenesis by phenotypic evaluation of miR-202 knockout (KO) mice. Loss of miR-202 results in spermatocyte apoptosis and perturbation of the zygonema-to-pachynema transition. Multiple processes during meiosis prophase I including synapsis and crossover formation are disrupted, and inter-sister chromatid synapses are detected. Moreover, we demonstrate that Separase mRNA is a miR-202 direct target and provides evidence that miR-202 upregulates REC8 by repressing Separase expression. Therefore, we have identified miR-202 as a new regulating noncoding gene that acts on the established SEPARASE-REC8 axis in meiosis.


Subject(s)
Cell Cycle Proteins , MicroRNAs , Separase , Animals , Cell Cycle Proteins/metabolism , Chromatids/metabolism , Male , Meiosis/genetics , Mice , MicroRNAs/genetics , Separase/genetics
9.
Sci Adv ; 8(21): eabn1606, 2022 May 27.
Article in English | MEDLINE | ID: mdl-35613276

ABSTRACT

The chromatin state, which undergoes global changes during spermatogenesis, is critical to meiotic initiation and progression. However, the key regulators involved and the underlying molecular mechanisms remain to be uncovered. Here, we report that mouse BEND2 is specifically expressed in spermatogenic cells around meiotic initiation and that it plays an essential role in meiotic progression. Bend2 gene knockout in male mice arrested meiosis at the transition from zygonema to pachynema, disrupted synapsis and DNA double-strand break repair, and induced nonhomologous chromosomal pairing. BEND2 interacted with chromatin-associated proteins that are components of certain transcription-repressor complexes. BEND2-binding sites were identified in diverse chromatin states and enriched in simple sequence repeats. BEND2 inhibited the expression of genes involved in meiotic initiation and regulated chromatin accessibility and the modification of H3K4me3. Therefore, our study identified BEND2 as a previously unknown key regulator of meiosis, gene expression, and chromatin state during mouse spermatogenesis.

10.
Development ; 148(24)2021 12 15.
Article in English | MEDLINE | ID: mdl-34913465

ABSTRACT

Spermatogonial differentiation and meiotic initiation during spermatogenesis are tightly regulated by a number of genes, including those encoding enzymes for miRNA biogenesis. However, whether and how single miRNAs regulate these processes remain unclear. Here, we report that miR-202, a member of the let-7 family, prevents precocious spermatogonial differentiation and meiotic initiation in spermatogenesis by regulating the timely expression of many genes, including those for key regulators such as STRA8 and DMRT6. In miR-202 knockout (KO) mice, the undifferentiated spermatogonial pool is reduced, accompanied by age-dependent decline of fertility. In KO mice, SYCP3, STRA8 and DMRT6 are expressed earlier than in wild-type littermates, and Dmrt6 mRNA is a direct target of miR-202-5p. Moreover, the precocious spermatogonial differentiation and meiotic initiation were also observed in KO spermatogonial stem cells when cultured and induced in vitro, and could be partially rescued by the knockdown of Dmrt6. Therefore, we have not only shown that miR-202 is a regulator of meiotic initiation but also identified a previously unknown module in the underlying regulatory network.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , MicroRNAs/genetics , Spermatogenesis/genetics , Spermatogonia/growth & development , Testis/growth & development , Adult Germline Stem Cells/cytology , Animals , Cell Cycle Proteins/genetics , Cell Differentiation/genetics , DNA-Binding Proteins/genetics , Fertility/genetics , Gene Expression Regulation, Developmental/genetics , Male , Meiosis/genetics , Mice , Mice, Knockout , Spermatogonia/metabolism , Testis/metabolism , Transcription Factors/genetics
11.
Front Cell Dev Biol ; 9: 673258, 2021.
Article in English | MEDLINE | ID: mdl-34113620

ABSTRACT

Protein arginine methyltransferase 5 (PRMT5) catalyzes the formation of mono- or symmetric dimethylarginine residues on histones and non-histone substrates and has been demonstrated to play important roles in many biological processes. In the present study, we observed that PRMT5 is abundantly expressed in spermatogonial stem cells (SSCs) and that Prmt5 deletion results in a progressive loss of SSCs and male infertility. The proliferation of Prmt5-deficient SSCs cultured in vitro exhibited abnormal proliferation, cell cycle arrest in G0/G1 phase and a significant increase in apoptosis. Furthermore, PLZF expression was dramatically reduced in Prmt5-deficient SSCs, and the levels of H3K9me2 and H3K27me2 were increased in the proximal promoter region of the Plzf gene in Prmt5-deficient SSCs. Further study revealed that the expression of lysine demethylases (JMJD1A, JMJD1B, JMJD1C, and KDM6B) was significantly reduced in Prmt5-deficient SSCs and that the level of permissive arginine methylation H3R2me2s was significantly decreased at the upstream promoter region of these genes in Prmt5-deficient SSCs. Our results demonstrate that PRMT5 regulates spermatogonial stem cell development by modulating histone H3 lysine modifications.

12.
Proc Natl Acad Sci U S A ; 118(8)2021 02 23.
Article in English | MEDLINE | ID: mdl-33602822

ABSTRACT

Meiosis is a specialized cell division that creates haploid germ cells from diploid progenitors. Through differential RNA expression analyses, we previously identified a number of mouse genes that were dramatically elevated in spermatocytes, relative to their very low expression in spermatogonia and somatic organs. Here, we investigated in detail 1700102P08Rik, one of these genes, and independently conclude that it encodes a male germline-specific protein, in agreement with a recent report. We demonstrated that it is essential for pachynema progression in spermatocytes and named it male pachynema-specific (MAPS) protein. Mice lacking Maps (Maps-/- ) suffered from pachytene arrest and spermatocyte death, leading to male infertility, whereas female fertility was not affected. Interestingly, pubertal Maps-/- spermatocytes were arrested at early pachytene stage, accompanied by defects in DNA double-strand break (DSB) repair, crossover formation, and XY body formation. In contrast, adult Maps-/- spermatocytes only exhibited partially defective crossover but nonetheless were delayed or failed in progression from early to mid- and late pachytene stage, resulting in cell death. Furthermore, we report a significant transcriptional dysregulation in autosomes and XY chromosomes in both pubertal and adult Maps-/- pachytene spermatocytes, including failed meiotic sex chromosome inactivation (MSCI). Further experiments revealed that MAPS overexpression in vitro dramatically decreased the ubiquitination levels of cellular proteins. Conversely, in Maps-/- pachytene cells, protein ubiquitination was dramatically increased, likely contributing to the large-scale disruption in gene expression in pachytene cells. Thus, MAPS is a protein essential for pachynema progression in male mice, possibly in mammals in general.


Subject(s)
Infertility, Male/pathology , Meiosis , Nuclear Proteins/physiology , Pachytene Stage , Spermatocytes/pathology , Spermatogenesis , Animals , Chromosome Pairing , DNA Repair , Female , Infertility, Male/etiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Sex Chromosomes , Spermatocytes/metabolism
13.
Sci Rep ; 11(1): 475, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33436645

ABSTRACT

Clostridium perfringens (Cp) is a ubiquitous opportunistic pathogen of humans and animals in the natural environment and animal intestines. The pathogenicity of Cp depends on the production of toxins encoded by genes on the chromosomes or plasmids. In contemporary literature, there is no clear consensus about the pathogenicity of CpA ß2 toxin. To analyze the homology of the genome of piglet source CpA and its ß2 toxin, we sequenced the whole genome of strain JXJA17 isolated from diarrhea piglets using the Illumina Miseq and Pacbio Sequel platforms. The genome was composed of a circular chromosome with 3,324,072 bp (G + C content: 28.51%) and nine plasmids. Genome and 16S rDNA homology analysis revealed a close relation of the JXJA17 strain with the JGS1495, Cp-06, Cp-16, and FORC_003 strains. These strains were isolated from different samples and belonged to different toxin-types. JXJA17 strain was found to carry two toxin genes (plc and cpb2). In contrast to other Cp strains, the cpb2 of JXJA17 was located on a large plasmid (58 kb) with no co-localization of other toxin genes or antibiotic resistance genes. Analysis of JXJA17 genome homology and its cpb2 would facilitate our further study the relationship between ß2 toxin and piglet diarrhea.


Subject(s)
Bacterial Toxins/genetics , Clostridium Infections/veterinary , Clostridium perfringens/genetics , Clostridium perfringens/isolation & purification , Swine Diseases/microbiology , Animals , China , Chromosome Mapping , Chromosomes , Clostridium Infections/genetics , Clostridium Infections/microbiology , Diarrhea/genetics , Diarrhea/microbiology , Drug Resistance, Bacterial/genetics , Genome, Bacterial , Plasmids , Sequence Analysis, DNA , Sequence Homology , Swine , Swine Diseases/genetics , Whole Genome Sequencing
15.
iScience ; 23(4): 101034, 2020 Apr 24.
Article in English | MEDLINE | ID: mdl-32315832

ABSTRACT

Three-dimensional chromatin structures undergo dynamic reorganization during mammalian spermatogenesis; however, their impacts on gene regulation remain unclear. Here, we focused on understanding the structure-function regulation of meiotic chromosomes by Hi-C and other omics techniques in mouse spermatogenesis across five stages. Beyond confirming recent reports regarding changes in compartmentalization and reorganization of topologically associating domains (TADs), we further demonstrated that chromatin loops are present prior to and after, but not at, the pachytene stage. By integrating Hi-C and RNA-seq data, we showed that the switching of A/B compartments between spermatogenic stages is tightly associated with meiosis-specific mRNAs and piRNAs expression. Moreover, our ATAC-seq data indicated that chromatin accessibility per se is not responsible for the TAD and loop diminishment at pachytene. Additionally, our ChIP-seq data demonstrated that CTCF and cohesin remain bound at TAD boundary regions throughout meiosis, suggesting that dynamic reorganization of TADs does not require CTCF and cohesin clearance.

17.
Sci Adv ; 5(8): eaax1101, 2019 08.
Article in English | MEDLINE | ID: mdl-31453335

ABSTRACT

Meiosis is a specialized type of cell division that creates haploid germ cells and ensures their genetic diversity through homologous recombination. We show that the H3K4me3 reader ZCWPW1 is specifically required for meiosis prophase I progression in male but not in female germ cells in mice. Loss of Zcwpw1 in male mice caused a complete failure of synapsis, resulting in meiotic arrest at the zygotene to pachytene stage, accompanied by incomplete DNA double-strand break repair and lack of crossover formation, leading to male infertility. In oocytes, deletion of Zcwpw1 only somewhat slowed down meiosis prophase I progression; Zcwpw1-/- oocytes were able to complete meiosis, and Zcwpw1-/- female mice had normal fertility until mid-adulthood. We conclude that the H3K4me3 reader ZCWPW1 is indispensable for meiosis synapsis in males but is dispensable for females. Our results suggest that ZCWPW1 may represent a previously unknown, sex-dependent epigenetic regulator of germ cell meiosis in mammals.


Subject(s)
Cell Cycle Proteins/physiology , DNA End-Joining Repair/genetics , Histone Code/genetics , Meiotic Prophase I/genetics , Oocytes/cytology , Spermatozoa/cytology , Animals , Cell Cycle Proteins/genetics , DNA Breaks, Double-Stranded , Female , Histones/genetics , Infertility, Male/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Sex Factors
18.
Biol Reprod ; 100(6): 1673-1685, 2019 06 01.
Article in English | MEDLINE | ID: mdl-31087039

ABSTRACT

Family with sequence similarity 46, member C (FAM46C) is a highly conserved non-canonical RNA polyadenylation polymerase that is abundantly expressed in human and mouse testes and is frequently mutated in patients with multiple myeloma. However, its physiological role remains largely unknown. In this study, we found that FAM46C is specifically localized to the manchette of spermatids in mouse testes, a transient microtubule-based structure mainly involved in nuclear shaping and intra-flagellar protein traffic. Gene knockout of FAM46C in mice resulted in male sterility, characterized by the production of headless spermatozoa in testes. Sperm heads were intermittently found in the epididymides of FAM46C knockout mice, but their fertilization ability was severely compromised based on the results of intracytoplasmic sperm injection assays. Interestingly, our RNA-sequencing analyses of FAM46C knockout testes revealed that mRNA levels of only nine genes were significantly altered compared to wild-type ones (q < 0.05). When considering alternate activities for FAM46C, in vitro assays demonstrated that FAM46C does not exhibit protein kinase or AMPylation activity against general substrates. Together, our data show that FAM46C in spermatids is a novel component in fastening the sperm head and flagellum.


Subject(s)
Flagella/physiology , Polynucleotide Adenylyltransferase/physiology , Sperm Head/physiology , Spermatids/physiology , Spermatogenesis/genetics , Animals , Cell Differentiation/genetics , Cells, Cultured , Female , Flagella/metabolism , Infertility, Male/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Polynucleotide Adenylyltransferase/genetics , Pregnancy , Sperm Head/metabolism , Spermatids/cytology , Spermatozoa/physiology
19.
Anaerobe ; 55: 136-141, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30529715

ABSTRACT

Though the problem of neonatal piglet diarrhea is well known, the differences in the bacterial diversity and community composition between healthy and diarrheal piglets are still unknown. We investigated these differences in neonatal piglets from Jiangxi Province, China. Healthy (H, n = 20) and diarrheal (D, n = 20) piglets were selected from six regions. The fecal microbial communities were analyzed by sequencing the V3V4 region of 16S rRNA gene. We found the ratio of major phyla (Fusobacteria, Bacteroidetes, Firmicutes and Proteobacteria) was >99% of 7 phyla. The overall alpha diversity indices, such as chao, sobs, coverage and Shannon, were not significantly different. Moreover, the relative abundance of the predicted functions was highly similar in the two groups. Our results indicated that Clostridium was divided into two major groups: Clostridium sensu stricto_1 and stricto_2. Sensu stricto_2 was highly abundant in the D group and low abundance in the H group, whereas the results of sensu stricto_1 were opposite. Comparative analyses within the H or D groups showed that Escherichia-Shigella and Streptococcus at the genus level and unclassified Lactobacillus at the species level were significant difference. Comparative analyses of the two groups showed that unclassified Prevotellaceae at the genus level and Fusobacterium mortifierum were significantly different and had high linear discriminant analysis (LDA) scores. The significantly different microbes composition results also existed in the same litter, based on excluding regions influence. These results suggested that piglet diarrhea was closely associated with these microbes. This study provides insights into gut microbial interactions and prevention of piglet diarrhea.


Subject(s)
Bacteria/classification , Diarrhea/veterinary , Gastrointestinal Microbiome , Swine Diseases/microbiology , Animals , Animals, Newborn , Bacteria/genetics , China , Cluster Analysis , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , DNA, Ribosomal/chemistry , DNA, Ribosomal/genetics , Diarrhea/microbiology , Genetic Variation , Phylogeny , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA , Swine
20.
Development ; 145(11)2018 05 30.
Article in English | MEDLINE | ID: mdl-29848638

ABSTRACT

The postmeiotic development of male germ cells, also known as spermiogenesis, features the coordinated expression of a large number of spermatid-specific genes. However, only a limited number of key transcription factors have been identified and the underlying regulatory mechanisms remain largely unknown. Here, we report that SOX30, the most-divergent member of the Sry-related high-motility group box (SOX) family of transcription factors, is essential for mouse spermiogenesis. The SOX30 protein was predominantly expressed in spermatids, while its transcription was regulated by retinoic acid and by MYBL1 before and during meiosis. Sox30 knockout mice arrested spermiogenesis at step 3 round spermatids, which underwent apoptosis and abnormal chromocenter formation. We also determined that SOX30 regulated the expression of hundreds of spermatid-specific protein-coding and long non-coding RNA genes. SOX30 bound to the proximal promoter of its own gene and activated its transcription. These results reveal SOX30 as a novel key regulator of spermiogenesis that regulates its own transcription to enforce and activate this meiotic regulatory pathway.


Subject(s)
Gene Expression Regulation/genetics , SOX Transcription Factors/genetics , Spermatids/metabolism , Spermatogenesis/physiology , Animals , Apoptosis/physiology , Male , Meiosis/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-myb/genetics , Trans-Activators/genetics , Tretinoin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...