Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
Add more filters










Publication year range
1.
Cells ; 13(9)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38727278

ABSTRACT

Spermatogenesis involves a complex process of cellular differentiation maintained by spermatogonial stem cells (SSCs). Being critical to male reproduction, it is generally assumed that spermatogenesis starts and ends in equivalent transcriptional states in related species. Based on single-cell gene expression profiling, it has been proposed that undifferentiated human spermatogonia can be subclassified into four heterogenous subtypes, termed states 0, 0A, 0B, and 1. To increase the resolution of the undifferentiated compartment and trace the origin of the spermatogenic trajectory, we re-analysed the single-cell (sc) RNA-sequencing libraries of 34 post-pubescent human testes to generate an integrated atlas of germ cell differentiation. We then used this atlas to perform comparative analyses of the putative SSC transcriptome both across human development (using 28 foetal and pre-pubertal scRNA-seq libraries) and across species (including data from sheep, pig, buffalo, rhesus and cynomolgus macaque, rat, and mouse). Alongside its detailed characterisation, we show that the transcriptional heterogeneity of the undifferentiated spermatogonial cell compartment varies not only between species but across development. Our findings associate 'state 0B' with a suppressive transcriptomic programme that, in adult humans, acts to functionally oppose proliferation and maintain cells in a ready-to-react state. Consistent with this conclusion, we show that human foetal germ cells-which are mitotically arrested-can be characterised solely as state 0B. While germ cells with a state 0B signature are also present in foetal mice (and are likely conserved at this stage throughout mammals), they are not maintained into adulthood. We conjecture that in rodents, the foetal-like state 0B differentiates at birth into the renewing SSC population, whereas in humans it is maintained as a reserve population, supporting testicular homeostasis over a longer reproductive lifespan while reducing mutagenic load. Together, these results suggest that SSCs adopt differing evolutionary strategies across species to ensure fertility and genome integrity over vastly differing life histories and reproductive timeframes.


Subject(s)
Spermatogonia , Humans , Animals , Male , Spermatogonia/cytology , Spermatogonia/metabolism , Adult Germline Stem Cells/metabolism , Adult Germline Stem Cells/cytology , Cell Differentiation/genetics , Spermatogenesis/genetics , Transcriptome/genetics , Adult , Mice , Fetus/cytology , Testis/cytology , Testis/metabolism , Rodentia , Rats , Single-Cell Analysis
2.
Cell Prolif ; 56(4): e13390, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36592615

ABSTRACT

Spermatogonial stem cell (SSC) self-renewal is regulated by reciprocal interactions between Sertoli cells and SSCs in the testis. In a previous study, microtubule-associated serine/threonine kinase 4 (MAST4) has been studied in Sertoli cells as a regulator of SSC self-renewal. The present study focused on the mechanism by which MAST4 in Sertoli cells transmits the signal and regulates SSCs, especially cell cycle regulation. The expression of PLZF, CDK2 and PLZF target genes was examined in WT and Mast4 KO testes by Immunohistochemistry, RT-qPCR and western blot. In addition, IdU and BrdU were injected into WT and Mast4 KO mice and cell cycle of SSCs was analysed. Finally, the testis tissues were cultured in vitro to examine the regulation of cell cycle by MAST4 pathway. Mast4 KO mice showed infertility with Sertoli cell-only syndrome and reduced sperm count. Furthermore, Mast4 deletion led to decreased PLZF expression and cell cycle progression in the testes. MAST4 also induced cyclin-dependent kinase 2 (CDK2) to phosphorylate PLZF and activated PLZF suppressed the transcriptional levels of genes related to cell cycle arrest, leading SSCs to remain stem cell state. MAST4 is essential for maintaining cell cycle in SSCs via the CDK2-PLZF interaction. These results demonstrate the pivotal role of MAST4 regulating cell cycle of SSCs and the significance of spermatogenesis.


Subject(s)
Adult Germline Stem Cells , Microtubule-Associated Proteins , Animals , Mice , Adult Germline Stem Cells/cytology , Adult Germline Stem Cells/physiology , Cell Cycle/physiology , Microtubule-Associated Proteins/physiology , Male
3.
Cell Prolif ; 55(3): e13195, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35119145

ABSTRACT

OBJECTIVES: Spermatogonial stem cells (SSCs), the germline stem cells (GSCs) committed to spermatogenesis in niche, can transform into pluripotent state in long-term culture without introduction of exogenous factors, typically in p53 deficiency condition. As the guardian for genomic stability, p53 is associated with epigenetic alterations during SSCs transformation. However, the mechanism is still unknown, since complicated roles of p53 baffle our understanding of the regulating process. MATERIALS AND METHODS: The chromatin accessibility and differentially expressed genes (DEGs) were analysed in p53+/+ and p53-/- SSCs using the Assay for Transposase-Accessible Chromatin with high-throughput Sequencing (ATAC-seq) and RNA-sequencing (RNA-seq), to explore the connection of p53 and cell fate at chromosomal level. RESULTS: Several transcription factors (TFs), such as CTCF, SMAD3 and SOX2, were predicted as important factors mediating the transformation. Molecular evidence suggested that SMAD3 efficiently promoted pluripotency-associated gene expression both in fresh and long-term cultured SSCs. However, p53 knockout (KO) is insufficient to induce SMAD3 expression in SSCs. CONCLUSIONS: These observations indicate that SMAD3 is a key factor for SSCs transformation, and an unknown event is required to activate SMAD3 as the prerequisite for SSCs reprogramming, which may occur in the long-term culture of SSCs. This study demonstrates the connection of p53 and pluripotency-associated factors, providing new insight for understanding the mechanisms of SSCs reprogramming and germline tumorigenesis.


Subject(s)
Adult Germline Stem Cells/cytology , Chromatin/metabolism , Pluripotent Stem Cells/cytology , Spermatogenesis/physiology , Tumor Suppressor Protein p53/deficiency , Animals , Cell Differentiation/physiology , Cellular Reprogramming/physiology , Male , Mice, Transgenic , Spermatogonia/metabolism , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism
4.
J Biol Chem ; 298(2): 101559, 2022 02.
Article in English | MEDLINE | ID: mdl-34979097

ABSTRACT

Spermatogonial stem cells (SSCs) are able to undergo both self-renewal and differentiation. Unlike self-renewal, which replenishes the SSC and progenitor pool, differentiation is an irreversible process committing cells to meiosis. Although the preparations for meiotic events in differentiating spermatogonia (Di-SG) are likely to be accompanied by alterations in chromatin structure, the three-dimensional chromatin architectural differences between SSCs and Di-SG, and the higher-order chromatin dynamics during spermatogonial differentiation, have not been systematically investigated. Here, we performed in situ high-throughput chromosome conformation capture, RNA-seq, and chromatin immunoprecipitation-sequencing analyses on porcine undifferentiated spermatogonia (which consist of SSCs and progenitors) and Di-SG. We identified that Di-SG exhibited less compact chromatin structural organization, weakened compartmentalization, and diminished topologically associating domains in comparison with undifferentiated spermatogonia, suggesting that diminished higher-order chromatin architecture in meiotic cells, as shown by recent reports, might be preprogrammed in Di-SG. Our data also revealed that A/B compartments, representing open or closed chromatin regions respectively, and topologically associating domains were related to dynamic gene expression during spermatogonial differentiation. Furthermore, we unraveled the contribution of promoter-enhancer interactions to premeiotic transcriptional regulation, which has not been accomplished in previous studies due to limited cell input and resolution. Together, our study uncovered the three-dimensional chromatin structure of SSCs/progenitors and Di-SG, as well as the interplay between higher-order chromatin architecture and dynamic gene expression during spermatogonial differentiation. These findings provide novel insights into the mechanisms for SSC self-renewal and differentiation and have implications for diagnosis and treatment of male sub-/infertility.


Subject(s)
Adult Germline Stem Cells , Chromatin , Spermatogenesis , Spermatogonia , Adult Germline Stem Cells/cytology , Adult Germline Stem Cells/metabolism , Animals , Cell Differentiation/physiology , Chromatin/metabolism , Male , Spermatogenesis/physiology , Spermatogonia/cytology , Swine
5.
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
6.
Open Biol ; 11(11): 210107, 2021 11.
Article in English | MEDLINE | ID: mdl-34727723

ABSTRACT

The centromere is the constricted chromosomal region required for the correct separation of the genetic material at cell division. The kinetochore protein complex assembles at the centromere and captures microtubules emanating from the centrosome to orchestrate chromosome segregation in mitosis and meiosis. Asymmetric cell division (ACD) is a special type of mitosis that generates two daughter cells with different fates. Epigenetic mechanisms operating at the centromere have been proposed to contribute to ACD. Recent studies have shown that an asymmetric distribution of CENP-A-the centromere-specific histone H3 variant-between sister chromatids can bias chromosome segregation in ACD. In stem cells, this leads to non-random sister chromatid segregation, which can affect cell fate. These findings support the 'silent sister' hypothesis, according to which the mechanisms of ACD are epigenetically regulated through centromeres. Here, we review the recent data implicating centromeres in ACDs and cell fate in Drosophila melanogaster female and male germline stem cells.


Subject(s)
Adult Germline Stem Cells/cytology , Centromere Protein A/metabolism , Centromere/metabolism , Drosophila Proteins/metabolism , Ovary/cytology , Adult Germline Stem Cells/metabolism , Animals , Asymmetric Cell Division , Drosophila melanogaster , Epigenesis, Genetic , Female , Male , Stem Cells/cytology , Stem Cells/metabolism
7.
Cells ; 10(7)2021 07 14.
Article in English | MEDLINE | ID: mdl-34359947

ABSTRACT

Male infertility is a major health problem affecting about 8-12% of couples worldwide. Spermatogenesis starts in the early fetus and completes after puberty, passing through different stages. Male infertility can result from primary or congenital, acquired, or idiopathic causes. The absence of sperm in semen, or azoospermia, results from non-obstructive causes (pretesticular and testicular), and post-testicular obstructive causes. Several medications such as antihypertensive drugs, antidepressants, chemotherapy, and radiotherapy could lead to impaired spermatogenesis and lead to a non-obstructive azoospermia. Spermatogonial stem cells (SSCs) are the basis for spermatogenesis and fertility in men. SSCs are characterized by their capacity to maintain the self-renewal process and differentiation into spermatozoa throughout the male reproductive life and transmit genetic information to the next generation. SSCs originate from gonocytes in the postnatal testis, which originate from long-lived primordial germ cells during embryonic development. The treatment of infertility in males has a poor prognosis. However, SSCs are viewed as a promising alternative for the regeneration of the impaired or damaged spermatogenesis. SSC transplantation is a promising technique for male infertility treatment and restoration of spermatogenesis in the case of degenerative diseases such as cancer, radiotherapy, and chemotherapy. The process involves isolation of SSCs and cryopreservation from a testicular biopsy before starting cancer treatment, followed by intra-testicular stem cell transplantation. In general, treatment for male infertility, even with SSC transplantation, still has several obstacles. The efficiency of cryopreservation, exclusion of malignant cells contamination in cancer patients, and socio-cultural attitudes remain major challenges to the wider application of SSCs as alternatives. Furthermore, there are limitations in experience and knowledge regarding cryopreservation of SSCs. However, the level of infrastructure or availability of regulatory approval to process and preserve testicular tissue makes them tangible and accurate therapy options for male infertility caused by non-obstructive azoospermia, though in their infancy, at least to date.


Subject(s)
Adult Germline Stem Cells/cytology , Azoospermia/drug therapy , Cell- and Tissue-Based Therapy , Infertility, Male/drug therapy , Spermatogenesis/drug effects , Cell- and Tissue-Based Therapy/methods , Humans , Male , Stem Cell Transplantation/methods
8.
PLoS Genet ; 17(7): e1009369, 2021 07.
Article in English | MEDLINE | ID: mdl-34237055

ABSTRACT

Spermatogonial stem cells (SSC), the foundation of spermatogenesis and male fertility, possess lifelong self-renewal activity. Aging leads to the decline in stem cell function and increased risk of paternal age-related genetic diseases. In the present study, we performed a comparative genomic analysis of mouse SSC-enriched undifferentiated spermatogonia (Oct4-GFP+/KIT-) and differentiating progenitors (Oct4-GFP+/KIT+) isolated from young and aged testes. Our transcriptome data revealed enormous complexity of expressed coding and non-coding RNAs and alternative splicing regulation during SSC differentiation. Further comparison between young and aged undifferentiated spermatogonia suggested these differentiation programs were affected by aging. We identified aberrant expression of genes associated with meiosis and TGF-ß signaling, alteration in alternative splicing regulation and differential expression of specific lncRNAs such as Fendrr. Epigenetic profiling revealed reduced H3K27me3 deposition at numerous pro-differentiation genes during SSC differentiation as well as aberrant H3K27me3 distribution at genes in Wnt and TGF-ß signaling upon aging. Finally, aged undifferentiated spermatogonia exhibited gene body hypomethylation, which is accompanied by an elevated 5hmC level. We believe this in-depth molecular analysis will serve as a reference for future analysis of SSC aging.


Subject(s)
Adult Germline Stem Cells/cytology , Adult Germline Stem Cells/physiology , Aging/physiology , Epigenome , 5-Methylcytosine/metabolism , Aging/genetics , Alternative Splicing , Animals , Cell Differentiation , Gene Expression Profiling , Gene Expression Regulation , Lysine/genetics , Lysine/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , RNA, Long Noncoding/genetics , Testis/cytology
9.
Development ; 148(10)2021 05 15.
Article in English | MEDLINE | ID: mdl-33998651

ABSTRACT

Heterochromatin-related epigenetic mechanisms, such as DNA methylation, facilitate pairing of homologous chromosomes during the meiotic prophase of mammalian spermatogenesis. In pro-spermatogonia, de novo DNA methylation plays a key role in completing meiotic prophase and initiating meiotic division. However, the role of maintenance DNA methylation in the regulation of meiosis, especially in the adult, is not well understood. Here, we reveal that NP95 (also known as UHRF1) and DNMT1 - two essential proteins for maintenance DNA methylation - are co-expressed in spermatogonia and are necessary for meiosis in male germ cells. We find that Np95- or Dnmt1-deficient spermatocytes exhibit spermatogenic defects characterized by synaptic failure during meiotic prophase. In addition, assembly of pericentric heterochromatin clusters in early meiotic prophase, a phenomenon that is required for subsequent pairing of homologous chromosomes, is disrupted in both mutants. Based on these observations, we propose that DNA methylation, established in pre-meiotic spermatogonia, regulates synapsis of homologous chromosomes and, in turn, quality control of male germ cells. Maintenance DNA methylation, therefore, plays a role in ensuring faithful transmission of both genetic and epigenetic information to offspring.


Subject(s)
CCAAT-Enhancer-Binding Proteins/genetics , Chromosome Pairing/genetics , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA Methylation/genetics , Spermatocytes/growth & development , Spermatogenesis/genetics , Ubiquitin-Protein Ligases/genetics , Adult Germline Stem Cells/cytology , Animals , CCAAT-Enhancer-Binding Proteins/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Epigenesis, Genetic/genetics , Heterochromatin/metabolism , Male , Mice , Mice, Knockout , Spermatocytes/physiology , Spermatogenesis/physiology , Ubiquitin-Protein Ligases/metabolism
11.
Reproduction ; 161(6): 645-655, 2021 05 05.
Article in English | MEDLINE | ID: mdl-33835049

ABSTRACT

Initiation of spermatogonial differentiation in the mouse testis begins with the response to retinoic acid (RA) characterized by activation of KIT and STRA8 expression. In the adult, spermatogonial differentiation is spatiotemporally coordinated by a pulse of RA every 8.6 days that is localized to stages VII-VIII of the seminiferous epithelial cycle. Dogmatically, progenitor spermatogonia that express retinoic acid receptor gamma (RARG) at these stages will differentiate in response to RA, but this has yet to be tested functionally. Previous single-cell RNA-seq data identified phenotypically and functionally distinct subsets of spermatogonial stem cells (SSCs) and progenitor spermatogonia, where late progenitor spermatogonia were defined by expression of RARG and Dppa3. Here, we found late progenitor spermatogonia (RARGhigh KIT-) were further divisible into two subpopulations based on Dppa3 reporter expression (Dppa3-ECFP or Dppa3-EGFP) and were observed across all stages of the seminiferous epithelial cycle. However, nearly all Dppa3+ spermatogonia were differentiating (KIT+) late in the seminiferous epithelial cycle (stages X-XII), while Dppa3- late progenitors remained abundant, suggesting that Dppa3+ and Dppa3- late progenitors differentially responded to RA. Following acute RA treatment (2-4 h), significantly more Dppa3+ late progenitors induced KIT, including at the midpoint of the cycle (stages VI-IX), than Dppa3- late progenitors. Subsequently, single-cell analyses indicated a subset of Dppa3+ late progenitors expressed higher levels of Rxra, which we confirmed by RXRA whole-mount immunostaining. Together, these results indicate RARG alone is insufficient to initiate a spermatogonial response to RA in the adult mouse testis and suggest differential RXRA expression may discriminate responding cells.


Subject(s)
Adult Germline Stem Cells/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Receptors, Retinoic Acid/metabolism , Retinoid X Receptor alpha/metabolism , Spermatogenesis , Spermatogonia/metabolism , Tretinoin/pharmacology , Adult Germline Stem Cells/cytology , Adult Germline Stem Cells/drug effects , Animals , Antineoplastic Agents/pharmacology , Chromosomal Proteins, Non-Histone/genetics , Male , Mice , Receptors, Retinoic Acid/genetics , Retinoid X Receptor alpha/genetics , Spermatogonia/cytology , Spermatogonia/drug effects , Retinoic Acid Receptor gamma
12.
Int J Mol Sci ; 22(4)2021 Feb 18.
Article in English | MEDLINE | ID: mdl-33670439

ABSTRACT

The spermatogonial stem cell (SSC) is a unique adult stem cell that requires tight physiological regulation during development and adulthood. As the foundation of spermatogenesis, SSCs are a potential tool for the treatment of infertility. Understanding the factors that are necessary for lifelong maintenance of a SSC pool in vivo is essential for successful in vitro expansion and safe downstream clinical usage. This review focused on the current knowledge of prepubertal testicular development and germ cell metabolism in different species, and implications for translational medicine. The significance of metabolism for cell biology, stem cell integrity, and fate decisions is discussed in general and in the context of SSC in vivo maintenance, differentiation, and in vitro expansion.


Subject(s)
Adult Germline Stem Cells/physiology , Cell Culture Techniques/methods , Cell Differentiation/physiology , Spermatogenesis/physiology , Spermatogonia/physiology , Adult , Adult Germline Stem Cells/cytology , Animals , Cells, Cultured , Humans , Male , Spermatogonia/cytology
13.
STAR Protoc ; 2(1): 100371, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33733240

ABSTRACT

The maintenance of stem cell populations and the differentiation of their progeny is coordinated by specific communication with associated niche cells. Here, we describe a protocol for short-term live imaging of the Drosophila ovarian germline stem cell niche ex vivo. By immobilizing the ovarian tissue in a fibrinogen-thrombin clot, we are able to maintain the tissue for short-term high-temporal live imaging. This enables the visualization of dynamic cellular processes, such as the cytoskeletal dynamics that control stem cell niche communication. For complete details on the use and execution of this protocol, please refer to Wilcockson and Ashe (2019).


Subject(s)
Adult Germline Stem Cells/cytology , Molecular Imaging/methods , Animals , Cell Differentiation , Drosophila/cytology , Female , Germ Cells/cytology , Oogonial Stem Cells/cytology , Ovary/cytology , Stem Cell Niche/physiology , Stem Cells/cytology
14.
Asian J Androl ; 23(3): 240-248, 2021.
Article in English | MEDLINE | ID: mdl-33533740

ABSTRACT

Spermatogonial stem cells (SSCs) have great applications in both reproductive and regenerative medicine. Primates including monkeys are very similar to humans with regard to physiology and pathology. Nevertheless, little is known about the isolation, the characteristics, and the culture of primate SSCs. This study was designed to identify, isolate, and culture monkey SSCs. Immunocytochemistry was used to identify markers for monkey SSCs. Glial cell line-derived neurotrophic factor family receptor alpha-1 (GFRA1)-enriched spermatogonia were isolated from monkeys, namely Macaca fascicularis (M. fascicularis), by two-step enzymatic digestion and magnetic-activated cell sorting, and they were cultured on precoated plates in the conditioned medium. Reverse transcription-polymerase chain reaction (RT-PCR), immunocytochemistry, and RNA sequencing were used to compare phenotype and transcriptomes in GFRA1-enriched spermatogonia between 0 day and 14 days of culture, and xenotransplantation was performed to evaluate the function of GFRA1-enriched spermatogonia. SSCs shared some phenotypes with rodent and human SSCs. GFRA1-enriched spermatogonia with high purity and viability were isolated from M. fascicularis testes. The freshly isolated cells expressed numerous markers for rodent SSCs, and they were cultured for 14 days. The expression of numerous SSC markers was maintained during the cultivation of GFRA1-enriched spermatogonia. RNA sequencing reflected a 97.3% similarity in global gene profiles between 0 day and 14 days of culture. The xenotransplantation assay indicated that the GFRA1-enriched spermatogonia formed colonies and proliferated in vivo in the recipient c-KitW/W (W) mutant mice. Collectively, GFRA1-enriched spermatogonia are monkey SSCs phenotypically both in vitro and in vivo. This study suggests that monkey might provide an alternative to human SSCs for basic research and application in human diseases.


Subject(s)
Adult Germline Stem Cells/cytology , Cell Separation/methods , Macaca fascicularis/classification , Analysis of Variance , Animals , Cell Separation/statistics & numerical data , Diabetes Complications , Disease Models, Animal , Humans , Rats, Sprague-Dawley
15.
Methods Mol Biol ; 2218: 37-47, 2021.
Article in English | MEDLINE | ID: mdl-33606221

ABSTRACT

Cryopreservation as a method that enables long-term storage of biological material has long been used for the conservation of valuable zebrafish genetic resources. However, currently, only spermatozoa of zebrafish can be successfully cryopreserved, while protocols for cryopreservation of eggs and embryos have not yet been fully developed. Transplantation of germline stem cells (GSCs) has risen as a favorable method that can bypass the current problem in cryopreservation of female genetic resources and can lead to reconstitution of fish species and lines through surrogate production. Here, we describe essential steps needed for the cryopreservation of spermatogonial stem cells (SSCs) and their utilization in the conservation of zebrafish genetic resources through SSC transplantation and surrogate production.


Subject(s)
Adult Germline Stem Cells/cytology , Cryopreservation/methods , Spermatogonia/cytology , Spermatozoa/cytology , Transplantation/methods , Adult Germline Stem Cells/drug effects , Animals , Cryoprotective Agents/pharmacology , Male , Spermatogonia/drug effects , Spermatozoa/drug effects , Testis/cytology , Testis/drug effects , Zebrafish/physiology
16.
Cell Rep ; 34(7): 108752, 2021 02 16.
Article in English | MEDLINE | ID: mdl-33596419

ABSTRACT

Spermatogonial stem cells (SSCs) sustain spermatogenesis by balancing self-renewal and initiation of differentiation to produce progenitor spermatogonia committed to forming sperm. To define the regulatory logic among SSCs and progenitors, we performed single-cell RNA velocity analyses and validated results in vivo. A predominant quiescent SSC population spawns a small subset of cell-cycle-activated SSCs via mitogen-activated protein kinase (MAPK)/AKT signaling. Activated SSCs form early progenitors and mTORC1 inhibition drives activated SSC accumulation consistent with blockade to progenitor formation. Mechanistically, mTORC1 inhibition suppresses transcription among spermatogonia and specifically alters expression of insulin growth factor (IGF) signaling in early progenitors. Tex14-/- testes lacking intercellular bridges do not accumulate activated SSCs following mTORC1 inhibition, indicating that steady-state mTORC1 signaling drives activated SSCs to produce progenitor clones. These results are consistent with a model of SSC self-renewal dependent on interconversion between activated and quiescent SSCs, and mTORC1-dependent initiation of differentiation from SSCs to progenitor clones.


Subject(s)
Adult Germline Stem Cells/physiology , Mechanistic Target of Rapamycin Complex 1/metabolism , Spermatogonia/physiology , Adult Germline Stem Cells/cytology , Adult Germline Stem Cells/metabolism , Animals , Cell Differentiation/physiology , Cells, Cultured , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction , Spermatogonia/metabolism
17.
Genes Dev ; 35(3-4): 250-260, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33446567

ABSTRACT

Reactive oxygen species (ROS) produced by NADPH1 oxidase 1 (NOX1) are thought to drive spermatogonial stem cell (SSC) self-renewal through feed-forward production of ROS by the ROS-BCL6B-NOX1 pathway. Here we report the critical role of oxygen on ROS-induced self-renewal. Cultured SSCs proliferated poorly and lacked BCL6B expression under hypoxia despite increase in mitochondria-derived ROS. Due to lack of ROS amplification under hypoxia, NOX1-derived ROS were significantly reduced, and Nox1-deficient SSCs proliferated poorly under hypoxia but normally under normoxia. NOX1-derived ROS also influenced hypoxic response in vivo because Nox1-deficient undifferentiated spermatogonia showed significantly reduced expression of HIF1A, a master transcription factor for hypoxic response. Hypoxia-induced poor proliferation occurred despite activation of MYC and suppression of CDKN1A by HIF1A, whose deficiency exacerbated self-renewal efficiency. Impaired proliferation of Nox1- or Hif1a-deficient SSCs under hypoxia was rescued by Cdkn1a depletion. Consistent with these observations, Cdkn1a-deficient SSCs proliferated actively only under hypoxia but not under normoxia. On the other hand, chemical suppression of mitochondria-derived ROS or Top1mt mitochondria-specific topoisomerase deficiency did not influence SSC fate, suggesting that NOX1-derived ROS play a more important role in SSCs than mitochondria-derived ROS. These results underscore the importance of ROS origin and oxygen tension on SSC self-renewal.


Subject(s)
Adult Germline Stem Cells/cytology , Cell Hypoxia/physiology , Oxygen/metabolism , Reactive Oxygen Species/metabolism , Animals , Cell Division/genetics , Cell Proliferation/genetics , Cells, Cultured , DNA Topoisomerases, Type I/genetics , Gene Expression Regulation, Developmental , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Mice , Mice, Knockout , Mitochondria/physiology , NADPH Oxidase 1/metabolism
18.
Biol Open ; 10(1)2021 01 06.
Article in English | MEDLINE | ID: mdl-33298570

ABSTRACT

Spermatogonial stem cell (SSC) transplantation is an alternative reproductive method to achieve conservation and production of elite animals in livestock production. Creating a recipient animal without endogenous germ cells is important for effective SSC transplantation. However, natural mutants with depletion of SSCs are difficult to obtain, and drug ablation of endogenous germ cells is arduous to perform for practical use. In this study, we used mouse models to study the preparation of recipients with congenital germ cell ablation. We knocked out (KO) Ets-variant gene 5 (Etv5) in mice using the CRISPR/Cas9 system. The testicular weight of Etv5-/- mice was significantly lower than that of wild-type (WT) mice. The germ cell layer of the seminiferous tubules gradually receded with age in Etv5-/- mice. At 12 weeks of age, the tubules of Etv5-/- mice lacked almost all spermatogenic cells with a Sertoli cell-only phenotype, and sperm were completely absent in the epididymis. We subsequently transplanted allogeneic SSCs with enhanced green fluorescent protein (EGFP) into 3- (immature) or 7-week-old (mature) Etv5-/- mice. Partial restoration of germ cell layers in the seminiferous tubules and spermatogenesis was observed in all immature testes but not in mature adult testes at 2 months post-transplantation. The presence of heterologous genes Etv5 and EGFP in recipient testicular tissue and epididymal sperm by PCR indicated that sperm originated from the transplanted donor cells. Our study demonstrates that, although Etv5-/- mice could accommodate and support foreign germ cell transplantation, this process occurs in a quite low efficiency to support a full spermatogenesis of transplanted SSCs. However, using Etv5-/- mice as a recipient model for SSC transplantation is feasible, and still needs further investigation to establish an optimized transplantation process.


Subject(s)
Adult Germline Stem Cells/cytology , Adult Germline Stem Cells/metabolism , DNA-Binding Proteins/genetics , Mice, Knockout , Models, Animal , Stem Cell Transplantation , Transcription Factors/genetics , Alleles , Animals , CRISPR-Cas Systems , DNA-Binding Proteins/deficiency , Gene Editing , Genotype , Mice , Transcription Factors/deficiency , Transplantation, Homologous
19.
Front Biosci (Landmark Ed) ; 26(1): 163-205, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33049667

ABSTRACT

Mammalian spermatogenesis is a complex but well-coordinated process in which spermatogonial stem cells (SSC) of the testis develop to form spermatozoa. During testicular homeostasis, the spermatogonial stem cells self-renew to maintain the stem cell pool or differentiate to form a progeny of germ cells which sequentially transform to spermatozoa. Accumulating evidence from clinical data and diverse model organisms suggest that the fate of spermatogonial stem cells towards self-renewal or differentiation is governed by intrinsic signals within the cells and by extracellular signals from the SSC niche. Here, we review the past and the most recent developments in understanding the nature of spermatogonial stem cells and the regulation of their homeostasis in mice. We also review the potential clinical applications of spermatogonial stem cells in male infertility as well as in germline modification, by virtue of gene correction and conversion of somatic cells to biologically competent male germline cells.


Subject(s)
Adult Germline Stem Cells/cytology , Cell Differentiation/physiology , Cell Self Renewal/physiology , Animals , Homeostasis , Humans , Male
20.
J Cell Physiol ; 236(2): 1391-1400, 2021 02.
Article in English | MEDLINE | ID: mdl-32749682

ABSTRACT

The development of primordial germ cells (PGCs) undergoes epigenetic modifications. The study of histone methylation in regulating PGCs is beneficial to understand the development and differentiation mechanism of germ stem cells. Notably, it provides a theoretical basis for directed induction and mass acquisition in vitro. However, little is known about the regulation of PGC formation by histone methylation. Here, we found the high enrichment of H3K4me2 in the blastoderm, genital ridges, and testis. Chromatin immunoprecipitation sequencing was performed and the results revealed that genomic H3K4me2 is dynamic in embryonic stem cells, PGCs, and spermatogonial stem cells. This trend was consistent with the H3K4me2 enrichment in the gene promoter region. Additionally, narrow region triggered PGC-related genes (Bmp4, Wnt5a, and Tcf7l2) and signaling pathways (Wnt and transforming growth factor-ß). After knocking down histone methylase Mll2 in vitro and vivo, the level of H3K4me2 decreased, inhibiting Cvh and Blimp1 expression, then repressing the formation of PGCs. Taken together, our study revealed the whole genome map of H3K4me2 in the formation of PGCs, contributing to improve the epigenetic study in PGC formation and providing materials for bird gene editing and rescue of endangered birds.


Subject(s)
Bone Morphogenetic Protein 4/genetics , Epigenesis, Genetic/genetics , Histone Methyltransferases/genetics , Testis/growth & development , Adult Germline Stem Cells/cytology , Adult Germline Stem Cells/metabolism , Animals , Blastoderm/growth & development , Cell Differentiation/genetics , Chickens/genetics , Chickens/growth & development , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental/genetics , Genitalia/growth & development , Germ Cells/growth & development , Male , Signal Transduction/genetics , Testis/metabolism , Transcription Factor 7-Like 2 Protein/genetics , Transforming Growth Factor beta/genetics , Wnt-5a Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...