Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.669
Filter
1.
J Biomed Mater Res B Appl Biomater ; 112(5): e35414, 2024 May.
Article in English | MEDLINE | ID: mdl-38733611

ABSTRACT

Utilizing natural scaffold production derived from extracellular matrix components presents a promising strategy for advancing in vitro spermatogenesis. In this study, we employed decellularized human placental tissue as a scaffold, upon which neonatal mouse spermatogonial cells (SCs) were cultured three-dimensional (3D) configuration. To assess cellular proliferation, we examined the expression of key markers (Id4 and Gfrα1) at both 1 and 14 days into the culture. Our quantitative reverse transcription-polymerase chain reaction (qRT-PCR) analysis revealed a notable increase in Gfrα1 gene expression, with the 3D culture group exhibiting the highest levels. Furthermore, the relative frequency of Gfrα1-positive cells significantly rose from 38.1% in isolated SCs to 46.13% and 76.93% in the two-dimensional (2D) and 3D culture systems, respectively. Moving forward to days 14 and 35 of the culture period, we evaluated the expression of differentiating markers (Sycp3, acrosin, and Protamine 1). Sycp3 and Prm1 gene expression levels were upregulated in both 2D and 3D cultures, with the 3D group displaying the highest expression. Additionally, acrosin gene expression increased notably within the 3D culture. Notably, at the 35-day mark, the percentage of Prm1-positive cells in the 3D group (36.4%) significantly surpassed that in the 2D group (10.96%). This study suggests that the utilization of placental scaffolds holds significant promise as a bio-scaffold for enhancing mouse in vitro spermatogenesis.


Subject(s)
Cell Differentiation , Cell Proliferation , Placenta , Animals , Female , Mice , Male , Humans , Placenta/cytology , Placenta/metabolism , Pregnancy , Spermatogonia/cytology , Spermatogonia/metabolism , Tissue Scaffolds/chemistry , Decellularized Extracellular Matrix/chemistry , Decellularized Extracellular Matrix/metabolism , Stem Cells/metabolism , Stem Cells/cytology
2.
Cell Mol Life Sci ; 81(1): 211, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38722330

ABSTRACT

Spermatogonial stem cells (SSCs) are capable of transmitting genetic information to the next generations and they are the initial cells for spermatogenesis. Nevertheless, it remains largely unknown about key genes and signaling pathways that regulate fate determinations of human SSCs and male infertility. In this study, we explored the expression, function, and mechanism of USP11 in controlling the proliferation and apoptosis of human SSCs as well as the association between its abnormality and azoospermia. We found that USP11 was predominantly expressed in human SSCs as shown by database analysis and immunohistochemistry. USP11 silencing led to decreases in proliferation and DNA synthesis and an enhancement in apoptosis of human SSCs. RNA-sequencing identified HOXC5 as a target of USP11 in human SSCs. Double immunofluorescence, Co-immunoprecipitation (Co-IP), and molecular docking demonstrated an interaction between USP11 and HOXC5 in human SSCs. HOXC5 knockdown suppressed the growth of human SSCs and increased apoptosis via the classical WNT/ß-catenin pathway. In contrast, HOXC5 overexpression reversed the effect of proliferation and apoptosis induced by USP11 silencing. Significantly, lower levels of USP11 expression were observed in the testicular tissues of patients with spermatogenic disorders. Collectively, these results implicate that USP11 regulates the fate decisions of human SSCs through the HOXC5/WNT/ß-catenin pathway. This study thus provides novel insights into understanding molecular mechanisms underlying human spermatogenesis and the etiology of azoospermia and it offers new targets for gene therapy of male infertility.


Subject(s)
Apoptosis , Cell Proliferation , Homeodomain Proteins , Wnt Signaling Pathway , Humans , Male , Apoptosis/genetics , Cell Proliferation/genetics , Wnt Signaling Pathway/genetics , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Azoospermia/metabolism , Azoospermia/genetics , Azoospermia/pathology , Spermatogonia/metabolism , Spermatogonia/cytology , Spermatogenesis/genetics , Adult Germline Stem Cells/metabolism , beta Catenin/metabolism , beta Catenin/genetics , Testis/metabolism , Testis/cytology , Thiolester Hydrolases
3.
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
4.
Int J Mol Sci ; 25(9)2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38732061

ABSTRACT

Embryonic stem-like cells (ES-like cells) are promising for medical research and clinical applications. Traditional methods involve "Yamanaka" transcription (OSKM) to derive these cells from somatic cells in vitro. Recently, a novel approach has emerged, obtaining ES-like cells from spermatogonia stem cells (SSCs) in a time-related process without adding artificial additives to cell cultures, like transcription factors or small molecules such as pten or p53 inhibitors. This study aims to investigate the role of the Nanog in the conversion of SSCs to pluripotent stem cells through both in silico analysis and in vitro experiments. We used bioinformatic methods and microarray data to find significant genes connected to this derivation path, to construct PPI networks, using enrichment analysis, and to construct miRNA-lncRNA networks, as well as in vitro experiments, immunostaining, and Fluidigm qPCR analysis to connect the dots of Nanog significance. We concluded that Nanog is one of the most crucial differentially expressed genes during SSC conversion, collaborating with critical regulators such as Sox2, Dazl, Pou5f1, Dnmt3, and Cdh1. This intricate protein network positions Nanog as a pivotal factor in pathway enrichment for generating ES-like cells, including Wnt signaling, focal adhesion, and PI3K-Akt-mTOR signaling. Nanog expression is presumed to play a vital role in deriving ES-like cells from SSCs in vitro. Finding its pivotal role in this path illuminates future research and clinical applications.


Subject(s)
Nanog Homeobox Protein , Nanog Homeobox Protein/metabolism , Nanog Homeobox Protein/genetics , Animals , Male , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/cytology , Cell Differentiation , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Spermatogonia/cytology , Spermatogonia/metabolism , Computer Simulation , Gene Regulatory Networks , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Gene Expression Profiling , Computational Biology/methods , Humans
5.
Cell Rep ; 43(4): 114113, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38625792

ABSTRACT

The continuous regeneration of spermatogonial stem cells (SSCs) underpins spermatogenesis and lifelong male fertility, but the developmental origins of the SSC pool remain unclear. Here, we document that hnRNPU is essential for establishing the SSC pool. In male mice, conditional loss of hnRNPU in prospermatogonia (ProSG) arrests spermatogenesis and results in sterility. hnRNPU-deficient ProSG fails to differentiate and migrate to the basement membrane to establish SSC pool in infancy. Moreover, hnRNPU deletion leads to the accumulation of ProSG and disrupts the process of T1-ProSG to T2-ProSG transition. Single-cell transcriptional analyses reveal that germ cells are in a mitotically quiescent state and lose their unique identity upon hnRNPU depletion. We further show that hnRNPU could bind to Vrk1, Slx4, and Dazl transcripts that have been identified to suffer aberrant alternative splicing in hnRNPU-deficient testes. These observations offer important insights into SSC pool establishment and may have translational implications for male fertility.


Subject(s)
Spermatogenesis , Spermatogonia , Animals , Male , Mice , Adult Germline Stem Cells/metabolism , Alternative Splicing/genetics , Cell Differentiation , Spermatogenesis/genetics , Spermatogonia/metabolism , Spermatogonia/cytology , Stem Cells/metabolism , Stem Cells/cytology , Testis/metabolism , Testis/cytology , Heterogeneous-Nuclear Ribonucleoprotein U/metabolism
6.
J Cell Physiol ; 239(4): e31202, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38291718

ABSTRACT

In the orchestrated environment of the testicular niche, the equilibrium between self-renewal and differentiation of spermatogonial stem cells (SSCs) is meticulously maintained, ensuring a stable stem cell reserve and robust spermatogenesis. Within this milieu, extracellular vesicles, specifically exosomes, have emerged as critical conveyors of intercellular communication. Despite their recognized significance, the implications of testicular exosomes in modulating SSC fate remain incompletely characterized. Given the fundamental support and regulatory influence of Sertoli cells (SCs) on SSCs, we were compelled to explore the role of SC-derived exosomes (SC-EXOs) in the SSC-testicular niche. Our investigation hinged on the hypothesis that SC-EXOs, secreted by SCs from the testes of 5-day-old mice-a developmental juncture marking the onset of SSC differentiation-participate in the regulation of this process. We discovered that exposure to SC-EXOs resulted in an upsurge of PLZF, MVH, and STRA8 expression in SSC cultures, concomitant with a diminution of ID4 and GFRA1 levels. Intriguingly, obstructing exosomal communication in a SC-SSC coculture system with the exosome inhibitor GW4869 attenuated SSC differentiation, suggesting that SC-EXOs may modulate this process via paracrine signaling. Further scrutiny revealed the presence of miR-493-5p within SC-EXOs, which suppresses Gdnf mRNA in SCs to indirectly restrain SSC differentiation through the modulation of GDNF expression-an indication of autocrine regulation. Collectively, our findings illuminate the complex regulatory schema by which SC-EXOs affect SSC differentiation, offering novel perspectives and laying the groundwork for future preclinical and clinical investigations.


Subject(s)
Autocrine Communication , Cell Differentiation , Exosomes , Paracrine Communication , Sertoli Cells , Spermatogonia , Animals , Male , Mice , Cell Differentiation/physiology , Exosomes/metabolism , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Mice, Inbred ICR , Sertoli Cells/cytology , Sertoli Cells/metabolism , Spermatogonia/cytology , Spermatogonia/metabolism
7.
Sheng Li Xue Bao ; 75(1): 17-26, 2023 Feb 25.
Article in Chinese | MEDLINE | ID: mdl-36859831

ABSTRACT

Previous studies have shown that long-term spermatogonial stem cells (SSCs) have the potential to spontaneously transform into pluripotent stem cells, which is speculated to be related to the tumorigenesis of testicular germ cells, especially when p53 is deficient in SSCs which shows a significant increase in the spontaneous transformation efficiency. Energy metabolism has been proved to be strongly associated with the maintenance and acquisition of pluripotency. Recently, we compared the difference in chromatin accessibility and gene expression profiles between wild-type (p53+/+) and p53 deficient (p53-/-) mouse SSCs using the Assay for Targeting Accessible-Chromatin with high-throughput sequencing (ATAC-seq) and transcriptome sequencing (RNA-seq) techniques, and revealed that SMAD3 is a key transcription factor in the transformation of SSCs into pluripotent cells. In addition, we also observed significant changes in the expression levels of many genes related to energy metabolism after p53 deletion. To further reveal the role of p53 in the regulation of pluripotency and energy metabolism, this paper explored the effects and mechanism of p53 deletion on energy metabolism during the pluripotent transformation of SSCs. The results of ATAC-seq and RNA-seq from p53+/+ and p53-/- SSCs revealed that gene chromatin accessibility related to positive regulation of glycolysis and electron transfer and ATP synthesis was increased, and the transcription levels of genes encoding key glycolytic enzymes and regulating electron transport-related enzymes were markedly increased. Furthermore, transcription factors SMAD3 and SMAD4 promoted glycolysis and energy homeostasis by binding to the chromatin of the Prkag2 gene which encodes the AMPK subunit. These results suggest that p53 deficiency activates the key enzyme genes of glycolysis in SSCs and enhances the chromatin accessibility of genes associated with glycolysis activation to improve glycolysis activity and promote transformation to pluripotency. Moreover, SMAD3/SMAD4-mediated transcription of the Prkag2 gene ensures the energy demand of cells in the process of pluripotency transformation and maintains cell energy homeostasis by promoting AMPK activity. These results shed light on the importance of the crosstalk between energy metabolism and stem cell pluripotency transformation, which might be helpful for clinical research of gonadal tumors.


Subject(s)
AMP-Activated Protein Kinases , Spermatogonia , Tumor Suppressor Protein p53 , Animals , Mice , Chromatin , Energy Metabolism , Gene Deletion , Stem Cells , Tumor Suppressor Protein p53/genetics , Spermatogonia/cytology , Male
8.
Nature ; 613(7943): 308-316, 2023 01.
Article in English | MEDLINE | ID: mdl-36544022

ABSTRACT

The testis produces gametes through spermatogenesis and evolves rapidly at both the morphological and molecular level in mammals1-6, probably owing to the evolutionary pressure on males to be reproductively successful7. However, the molecular evolution of individual spermatogenic cell types across mammals remains largely uncharacterized. Here we report evolutionary analyses of single-nucleus transcriptome data for testes from 11 species that cover the three main mammalian lineages (eutherians, marsupials and monotremes) and birds (the evolutionary outgroup), and include seven primates. We find that the rapid evolution of the testis was driven by accelerated fixation rates of gene expression changes, amino acid substitutions and new genes in late spermatogenic stages, probably facilitated by reduced pleiotropic constraints, haploid selection and transcriptionally permissive chromatin. We identify temporal expression changes of individual genes across species and conserved expression programs controlling ancestral spermatogenic processes. Genes predominantly expressed in spermatogonia (germ cells fuelling spermatogenesis) and Sertoli (somatic support) cells accumulated on X chromosomes during evolution, presumably owing to male-beneficial selective forces. Further work identified transcriptomal differences between X- and Y-bearing spermatids and uncovered that meiotic sex-chromosome inactivation (MSCI) also occurs in monotremes and hence is common to mammalian sex-chromosome systems. Thus, the mechanism of meiotic silencing of unsynapsed chromatin, which underlies MSCI, is an ancestral mammalian feature. Our study illuminates the molecular evolution of spermatogenesis and associated selective forces, and provides a resource for investigating the biology of the testis across mammals.


Subject(s)
Evolution, Molecular , Mammals , Spermatogenesis , Testis , Animals , Male , Chromatin/genetics , Mammals/genetics , Meiosis/genetics , Spermatogenesis/genetics , Testis/cytology , Transcriptome , Single-Cell Analysis , Birds/genetics , Primates/genetics , Gene Expression Regulation , Spermatogonia/cytology , Sertoli Cells/cytology , X Chromosome/genetics , Y Chromosome/genetics , Dosage Compensation, Genetic , Gene Silencing
9.
Asian J Androl ; 25(3): 322-330, 2023.
Article in English | MEDLINE | ID: mdl-36018067

ABSTRACT

Continuous self-renewal and differentiation of spermatogonial stem cells (SSCs) is vital for maintenance of adult spermatogenesis. Although several spermatogonial stem cell regulators have been extensively investigated in rodents, regulatory mechanisms of human SSC self-renewal and differentiation have not been fully established. We analyzed single-cell sequencing data from the human testis and found that forkhead box P4 (FOXP4) expression gradually increased with development of SSCs. Further analysis of its expression patterns in human testicular tissues revealed that FOXP4 specifically marks a subset of spermatogonia with stem cell potential. Conditional inactivation of FOXP4 in human SSC lines suppressed SSC proliferation and significantly activated apoptosis. FOXP4 expressions were markedly suppressed in tissues with dysregulated spermatogenesis. These findings imply that FOXP4 is involved in human SSC proliferation, which will help elucidate on the mechanisms controlling the fate decisions in human SSCs.


Subject(s)
Forkhead Transcription Factors , Spermatogonia , Adult , Humans , Male , Cell Differentiation , Cell Proliferation , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Spermatogenesis/genetics , Spermatogonia/cytology , Spermatogonia/metabolism , Stem Cells/metabolism , Testis/metabolism
10.
Acta Physiologica Sinica ; (6): 17-26, 2023.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-970102

ABSTRACT

Previous studies have shown that long-term spermatogonial stem cells (SSCs) have the potential to spontaneously transform into pluripotent stem cells, which is speculated to be related to the tumorigenesis of testicular germ cells, especially when p53 is deficient in SSCs which shows a significant increase in the spontaneous transformation efficiency. Energy metabolism has been proved to be strongly associated with the maintenance and acquisition of pluripotency. Recently, we compared the difference in chromatin accessibility and gene expression profiles between wild-type (p53+/+) and p53 deficient (p53-/-) mouse SSCs using the Assay for Targeting Accessible-Chromatin with high-throughput sequencing (ATAC-seq) and transcriptome sequencing (RNA-seq) techniques, and revealed that SMAD3 is a key transcription factor in the transformation of SSCs into pluripotent cells. In addition, we also observed significant changes in the expression levels of many genes related to energy metabolism after p53 deletion. To further reveal the role of p53 in the regulation of pluripotency and energy metabolism, this paper explored the effects and mechanism of p53 deletion on energy metabolism during the pluripotent transformation of SSCs. The results of ATAC-seq and RNA-seq from p53+/+ and p53-/- SSCs revealed that gene chromatin accessibility related to positive regulation of glycolysis and electron transfer and ATP synthesis was increased, and the transcription levels of genes encoding key glycolytic enzymes and regulating electron transport-related enzymes were markedly increased. Furthermore, transcription factors SMAD3 and SMAD4 promoted glycolysis and energy homeostasis by binding to the chromatin of the Prkag2 gene which encodes the AMPK subunit. These results suggest that p53 deficiency activates the key enzyme genes of glycolysis in SSCs and enhances the chromatin accessibility of genes associated with glycolysis activation to improve glycolysis activity and promote transformation to pluripotency. Moreover, SMAD3/SMAD4-mediated transcription of the Prkag2 gene ensures the energy demand of cells in the process of pluripotency transformation and maintains cell energy homeostasis by promoting AMPK activity. These results shed light on the importance of the crosstalk between energy metabolism and stem cell pluripotency transformation, which might be helpful for clinical research of gonadal tumors.


Subject(s)
Animals , Mice , Male , AMP-Activated Protein Kinases , Chromatin , Energy Metabolism , Gene Deletion , Stem Cells , Tumor Suppressor Protein p53/genetics , Spermatogonia/cytology
11.
Genes Dev ; 36(11-12): 752-763, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35738678

ABSTRACT

Self-renewal of spermatogonial stem cells is vital to lifelong production of male gametes and thus fertility. However, the underlying mechanisms remain enigmatic. Here, we show that DOT1L, the sole H3K79 methyltransferase, is required for spermatogonial stem cell self-renewal. Mice lacking DOT1L fail to maintain spermatogonial stem cells, characterized by a sequential loss of germ cells from spermatogonia to spermatids and ultimately a Sertoli cell only syndrome. Inhibition of DOT1L reduces the stem cell activity after transplantation. DOT1L promotes expression of the fate-determining HoxC transcription factors in spermatogonial stem cells. Furthermore, H3K79me2 accumulates at HoxC9 and HoxC10 genes. Our findings identify an essential function for DOT1L in adult stem cells and provide an epigenetic paradigm for regulation of spermatogonial stem cells.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Spermatogonia , Stem Cells , Animals , Cell Differentiation , Male , Mice , Spermatogonia/cytology , Spermatogonia/metabolism , Stem Cells/cytology , Stem Cells/metabolism
12.
Gene ; 823: 146390, 2022 May 20.
Article in English | MEDLINE | ID: mdl-35248658

ABSTRACT

Male fertility relies on continual and robust spermatogenesis. Environmental hypoxia adversely affects reproductive health in humans and animal studies provide compelling evidences that hypoxia impairs spermatogenesis in directly exposed individuals. However, a detail examination of hypoxia induced changes in testicular gene expression is still lacking and spermatogenesis in offspring of hypoxia exposed animals of awaits investigation. In this study, a hypobaric hypoxic chamber was used to simulate hypoxic conditions in mice and effects of hypoxia on spermatogenesis, fertility and testicular gene expression were evaluated. The results showed that hypoxia exposure reduced the number of undifferentiated spermatogonia but did not change the regenerative capacity of spermatogonial stem cells (SSCs) after transplantation. Hypoxia significantly increased the percent of abnormal sperm and these defects were recovered 2 months after returning to the normoxia. Transcriptome analysis of testicular tissues from control and hypoxia treated animals revealed that 766 genes were up-regulated and 965 genes were down-regulated. Surprisingly, expressions of genes that regulate epigenetic modifications were altered, indicating hypoxia-induced damage to spermatogenesis may be intergenerational. Indeed, animals that were sired by hypoxia exposed males exhibited impaired spermatogenesis. Together, these findings suggest that hypoxia exposure alters testicular gene expression and causes long-lasting damage to spermatogenesis.


Subject(s)
Gene Expression Profiling/methods , Gene Regulatory Networks , Hypoxia/genetics , Testis/chemistry , Animals , Epigenesis, Genetic , Gene Expression Regulation , Male , Mice , Spermatogenesis , Spermatogonia/cytology , Spermatogonia/transplantation , Testis/cytology
13.
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
14.
Andrology ; 10(2): 340-353, 2022 02.
Article in English | MEDLINE | ID: mdl-34499811

ABSTRACT

BACKGROUND: Cryopreservation can expand the usefulness of spermatogonial stem cells (SSCs) in various fields. However, previous investigations that have attempted to modulate cryoinjury-induced mechanisms to increase cryoprotective efficiency have mainly focused on apoptosis and necrosis. OBJECTIVES: This study aimed to establish an effective molecular-based cryoprotectant for SSC cryopreservation via autophagy modulation. MATERIALS AND METHODS: To determine the efficacy of autophagy modulation, we assessed the recovery rate and relative proliferation rate and performed western blotting for the determination of autophagy flux, immunocytochemistry and real-time quantitative polymerase chain reaction (RT-qPCR) for SSC characterization, and spermatogonial transplantation for in vivo SSC functional activity. RESULTS: The results showed that a basal level of autophagy caused a higher relative proliferation rate (pifithrin-µ 0.01 µM, 184.2 ± 11.2%; 3-methyladenine 0.01 µM, 175.3 ± 10.3%; pifithrin-µ 0.01 µM + 3-methyladenine 0.01 µM, P3, 224.6 ± 22.3%) than the DMSO control (100 ± 6.2%). All treatment groups exhibited normal characteristics, suggesting that these modulators could be used as effective cryoprotectants without changing the properties of the undifferentiated germ cells. According to the results of the in vivo spermatogonial transplantation assay, the colonies per total number of cultured SSCs was significantly higher in the pifithrin-µ 0.01 µM (1596.7 ± 172.5 colonies), 3-methyladenine 0.01 µM (1522.1 ± 179.2 colonies), and P3 (1727.5 ± 196.5 colonies) treatment groups than in the DMSO control (842.8 ± 110.08 colonies), which was comparable to that of the fresh control (1882.1 ± 132.1 colonies). DISCUSSION: A basal level of autophagy is more essential for resilience in frozen SSCs after thawing, rather than the excessive activation or inhibition of autophagy. CONCLUSION: A basal level of autophagy plays a critical role in the pro-survival response of frozen SSCs after thawing; herein, a new approach by which SSC cryoprotective efficiency can be improved was identified.


Subject(s)
Adult Germline Stem Cells/drug effects , Autophagy/drug effects , Cryopreservation , Cryoprotective Agents/pharmacology , Spermatogonia/cytology , Animals , Male , Mice
15.
Neuropeptides ; 91: 102215, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34883413

ABSTRACT

QRFP, an orexigenic neuropeptide, binds to its cognate receptor GPR103 and regulates various biological functions. We have recently shown that QRFP and its receptor are present in mice testes and that their expression is high during early postnatal period. The present study aimed to investigate the effect of sustained high level of QRFP on Sertoli cells proliferation and differentiation and to relate these events with germ cell differentiation and lumen formation in the seminiferous tubules in mice testes during prepubertal period. QRFP was injected intraperitoneally to male mice from postnatal day 5 to 16. Morphometric analysis and various markers related to Sertoli cell maturation (WT1, p27kip1, AMH, AR and CYP19A1) and germ cell proliferation and differentiation (PCNA, GDNF and c-Kit) were evaluated. QRFP administration caused an early lumen formation in the seminiferous tubules in testis of treated mice. Further, there was a significant increase in p27kip1 expression and a marked decrease in AMH expression in QRFP-treated mice compared to controls. However, no appreciable change was noted in AR expression in treated mice. QRFP treatment also caused an increase in c-Kit expression in treated mice compared to controls, suggesting an accelerated spermatogonial differentiation in testis of QRFP-treated mice. Taken together, the present results suggest that the prolonged high level of QRFP increases Sertoli cell maturation, which, in turn, plays a contributory role in increasing the pace of germ cell differentiation and formation of lumen in the seminiferous tubules.


Subject(s)
Neuropeptides/metabolism , Receptors, G-Protein-Coupled/metabolism , Sertoli Cells/metabolism , Testis/metabolism , Animals , Cell Proliferation/physiology , Male , Mice , Sertoli Cells/cytology , Spermatogonia/cytology , Spermatogonia/metabolism , Testis/cytology , Testis/growth & development
16.
Sci Rep ; 11(1): 24199, 2021 12 17.
Article in English | MEDLINE | ID: mdl-34921203

ABSTRACT

Germline mutations underlie genetic diversity and species evolution. Previous studies have assessed the theoretical mutation rates and spectra in germ cells mostly by analyzing genetic markers and reporter genes in populations and pedigrees. This study reported the direct measurement of germline mutations by whole-genome sequencing of cultured spermatogonial stem cells in mice, namely germline stem (GS) cells, together with multipotent GS (mGS) cells that spontaneously dedifferentiated from GS cells. GS cells produce functional sperm that can generate offspring by transplantation into seminiferous tubules, whereas mGS cells contribute to germline chimeras by microinjection into blastocysts in a manner similar to embryonic stem cells. The estimated mutation rate of GS and mGS cells was approximately 0.22 × 10-9 and 1.0 × 10-9 per base per cell population doubling, respectively, indicating that GS cells have a lower mutation rate compared to mGS cells. GS and mGS cells also showed distinct mutation patterns, with C-to-T transition as the most frequent in GS cells and C-to-A transversion as the most predominant in mGS cells. By karyotype analysis, GS cells showed recurrent trisomy of chromosomes 15 and 16, whereas mGS cells frequently exhibited chromosomes 1, 6, 8, and 11 amplifications, suggesting that distinct chromosomal abnormalities confer a selective growth advantage for each cell type in vitro. These data provide the basis for studying germline mutations and a foundation for the future utilization of GS cells for reproductive technology and clinical applications.


Subject(s)
Embryonic Stem Cells/metabolism , Genomic Instability/physiology , Animals , Chimera/metabolism , Computational Biology , Embryonic Stem Cells/cytology , Gene Expression Regulation, Developmental , Male , Mice , Mutation , Reactive Oxygen Species/metabolism , Seminiferous Tubules/metabolism , Spermatogonia/cytology , Spermatozoa
17.
Int J Mol Sci ; 22(23)2021 Nov 27.
Article in English | MEDLINE | ID: mdl-34884637

ABSTRACT

The adverse effects of radiation are proportional to the total dose and dose rate. We aimed to investigate the effects of radiation dose rate on different organs in mice. The mice were subjected to low dose rate (LDR, ~3.4 mGy/h) and high dose rate (HDR, ~51 Gy/h) radiation. LDR radiation caused severe tissue toxicity, as observed in the histological analysis of testis. It adversely influenced sperm production, including sperm count and motility, and induced greater sperm abnormalities. The expression of markers of early stage spermatogonial stem cells, such as Plzf, c-Kit, and Oct4, decreased significantly after LDR irradiation, compared to that following exposure of HDR radiation, in qPCR analysis. The compositional ratios of all stages of spermatogonia and meiotic cells, except round spermatid, were considerably reduced by LDR in FACS analysis. Therefore, LDR radiation caused more adverse testicular damage than that by HDR radiation, contrary to the response observed in other organs. Therefore, the dose rate of radiation may have differential effects, depending on the organ; it is necessary to evaluate the effect of radiation in terms of radiation dose, dose rate, organ type, and other conditions.


Subject(s)
Spermatogenesis/radiation effects , Testis/radiation effects , Animals , Dose-Response Relationship, Radiation , Gamma Rays , Male , Mice , Models, Animal , Radiation Dosage , Spermatids/cytology , Spermatids/radiation effects , Spermatogonia/cytology , Spermatogonia/radiation effects , Spermatozoa/cytology , Spermatozoa/radiation effects , Testis/cytology
18.
Sci Rep ; 11(1): 23007, 2021 11 26.
Article in English | MEDLINE | ID: mdl-34837027

ABSTRACT

Spermatogenesis, which is a continuous process from undifferentiated spermatogonia to spermatozoa in the seminiferous tubules, declines with age. To investigate changes in spermatogenesis with aging, we reconstructed the seminiferous tubules of 12 mice aged 12 to 30 months from serial sections and examined age-related and region-specific alterations in the seminiferous epithelium and spermatogenic waves in three dimensions. The basic structure of the seminiferous tubules, including the numbers of tubules, terminating points, branching points, and total tubule length, did not change with age. Age-related alterations in spermatogenesis, primarily assessed by the formation of vacuoles in Sertoli cells, were detected in the seminiferous tubules at 12 months. The proportion of altered tubule segments with impaired spermatogenesis further increased by 24 months, but remained unchanged thereafter. Altered tubule segments were preferentially distributed in tubule areas close to the rete testis and those in the center of the testis. Spermatogenic waves became shorter in length with age. These results provide a basis for examining the decline of spermatogenesis not only with aging, but also in male infertility.


Subject(s)
Aging , Seminiferous Tubules/ultrastructure , Spermatogenesis , Testis/ultrastructure , Animals , Male , Mice , Mice, Inbred C57BL , Seminiferous Epithelium/cytology , Seminiferous Epithelium/ultrastructure , Seminiferous Tubules/cytology , Spermatogonia/cytology , Spermatogonia/ultrastructure , Testis/cytology
19.
J Assist Reprod Genet ; 38(12): 3155-3173, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34661801

ABSTRACT

PURPOSE: Spermatogonial stem cells (SSCs) are the source for the mature male gamete. SSC technology in humans is mainly focusing on preserving fertility in cancer patients. Whereas in livestock, it is used for mining the factors associated with male fertility. The review discusses the present status of SSC biology, methodologies developed for in vitro culture, and challenges ahead in establishing SSC technology for the propagation of superior germplasm with special reference to livestock. METHOD: Published literatures from PubMed and Google Scholar on topics of SSCs isolation, purification, characterization, short and long-term culture of SSCs, stemness maintenance, epigenetic modifications of SSCs, growth factors, and SSC cryopreservation and transplantation were used for the study. RESULT: The fine-tuning of SSC isolation and culture conditions with special reference to feeder cells, growth factors, and additives need to be refined for livestock. An insight into the molecular mechanisms involved in maintaining stemness and proliferation of SSCs could facilitate the dissemination of superior germplasm through transplantation and transgenesis. The epigenetic influence on the composition and expression of the biomolecules during in vitro differentiation of cultured cells is essential for sustaining fertility. The development of surrogate males through gene-editing will be historic achievement for the foothold of the SSCs technology. CONCLUSION: Detailed studies on the species-specific factors regulating the stemness and differentiation of the SSCs are required for the development of a long-term culture system and in vitro spermatogenesis in livestock. Epigenetic changes in the SSCs during in vitro culture have to be elucidated for the successful application of SSCs for improving the productivity of the animals.


Subject(s)
Cell Culture Techniques/methods , Cell Transplantation/methods , Livestock/physiology , Spermatogonia/cytology , Spermatogonia/physiology , Stem Cells/cytology , Stem Cells/physiology , Adult Germline Stem Cells , Animals , Fertility , In Vitro Techniques/methods , Male , Spermatogenesis
20.
Stem Cell Reports ; 16(11): 2798-2812, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34653405

ABSTRACT

We propose a new concept that human somatic cells can be converted to become male germline stem cells by the defined factors. Here, we demonstrated that the overexpression of DAZL, DAZ2, and BOULE could directly reprogram human Sertoli cells into cells with the characteristics of human spermatogonial stem cells (SSCs), as shown by their similar transcriptomes and proteomics with human SSCs. Significantly, human SSCs derived from human Sertoli cells colonized and proliferated in vivo, and they could differentiate into spermatocytes and haploid spermatids in vitro. Human Sertoli cell-derived SSCs excluded Y chromosome microdeletions and assumed normal chromosomes. Collectively, human somatic cells could be converted directly to human SSCs with the self-renewal and differentiation potentials and high safety. This study is of unusual significance, because it provides an effective approach for reprogramming human somatic cells into male germ cells and offers invaluable male gametes for treating male infertility.


Subject(s)
Cell Differentiation/genetics , Cell Self Renewal/genetics , Cellular Reprogramming/genetics , RNA-Binding Proteins/genetics , Sertoli Cells/metabolism , Spermatogonia/metabolism , Animals , Cells, Cultured , Gene Expression Profiling/methods , Haploidy , Humans , Male , Mice, Nude , Proteomics/methods , RNA-Binding Proteins/metabolism , Sertoli Cells/cytology , Spermatids/cytology , Spermatids/metabolism , Spermatogonia/cytology , Stem Cell Transplantation/methods , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...