Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.853
Filter
1.
Zool Res ; 45(5): 1048-1060, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39147719

ABSTRACT

Extracellular membrane proteins are crucial for mediating cell attachment, recognition, and signal transduction in the testicular microenvironment, particularly germline stem cells. Cadherin 18 (CDH18), a type II classical cadherin, is primarily expressed in the nervous and reproductive systems. Here, we investigated the expression of CDH18 in neonatal porcine prospermatogonia (ProSGs) and murine spermatogonial stem cells (SSCs). Disruption of CDH18 expression did not adversely affect cell morphology, proliferation, self-renewal, or differentiation in cultured porcine ProSGs, but enhanced cell adhesion and prolonged cell maintenance. Transcriptomic analysis indicated that the down-regulation of CDH18 in ProSGs significantly up-regulated genes and signaling pathways associated with cell adhesion. To further elucidate the function of CDH18 in germ cells, Cdh18 knockout mice were generated, which exhibited normal testicular morphology, histology, and spermatogenesis. Transcriptomic analysis showed increased expression of genes associated with adhesion, consistent with the observations in porcine ProSGs. The interaction of CDH18 with ß-catenin and JAK2 in both porcine ProSGs and murine SSCs suggested an inhibitory effect on the canonical Wnt and JAK-STAT signaling pathways during CDH18 deficiency. Collectively, these findings highlight the crucial role of CDH18 in regulating cell adhesion in porcine ProSGs and mouse SSCs. Understanding this regulatory mechanism provides significant insights into the testicular niche.


Subject(s)
Cadherins , Cell Adhesion , Animals , Male , Swine , Cell Adhesion/physiology , Mice , Cadherins/metabolism , Cadherins/genetics , Mice, Knockout , Spermatogonia/metabolism , Spermatogonia/physiology , Testis/metabolism , Testis/physiology , Adult Germline Stem Cells/metabolism , Adult Germline Stem Cells/physiology , Gene Expression Regulation , Stem Cells/physiology , Stem Cells/metabolism
2.
Nat Commun ; 15(1): 6637, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39122675

ABSTRACT

piRNAs are crucial for transposon silencing, germ cell maturation, and fertility in male mice. Here, we report on the genetic landscape of piRNA dysfunction in humans and present 39 infertile men carrying biallelic variants in 14 different piRNA pathway genes, including PIWIL1, GTSF1, GPAT2, MAEL, TDRD1, and DDX4. In some affected men, the testicular phenotypes differ from those of the respective knockout mice and range from complete germ cell loss to the production of a few morphologically abnormal sperm. A reduced number of pachytene piRNAs was detected in the testicular tissue of variant carriers, demonstrating impaired piRNA biogenesis. Furthermore, LINE1 expression in spermatogonia links impaired piRNA biogenesis to transposon de-silencing and serves to classify variants as functionally relevant. These results establish the disrupted piRNA pathway as a major cause of human spermatogenic failure and provide insights into transposon silencing in human male germ cells.


Subject(s)
DNA Transposable Elements , Infertility, Male , RNA, Small Interfering , Spermatogenesis , Testis , Male , Humans , Spermatogenesis/genetics , Infertility, Male/genetics , Infertility, Male/metabolism , Infertility, Male/pathology , RNA, Small Interfering/metabolism , RNA, Small Interfering/genetics , DNA Transposable Elements/genetics , Animals , Testis/metabolism , Mice , Adult , Gene Silencing , Mice, Knockout , Argonaute Proteins/metabolism , Argonaute Proteins/genetics , Long Interspersed Nucleotide Elements/genetics , Spermatogonia/metabolism , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Piwi-Interacting RNA
3.
Int J Mol Sci ; 25(13)2024 Jun 25.
Article in English | MEDLINE | ID: mdl-39000031

ABSTRACT

In vitro maturation (IVM) is a promising fertility restoration strategy for patients with nonobstructive azoospermia or for prepubertal boys to obtain fertilizing-competent spermatozoa. However, in vitro spermatogenesis is still not achieved with human immature testicular tissue. Knowledge of various human testicular transcriptional profiles from different developmental periods helps us to better understand the testis development. This scoping review aims to describe the testis development and maturation from the fetal period towards adulthood and to find information to optimize IVM. Research papers related to native and in vitro cultured human testicular cells and single-cell RNA-sequencing (scRNA-seq) were identified and critically reviewed. Special focus was given to gene ontology terms to facilitate the interpretation of the biological function of related genes. The different consecutive maturation states of both the germ and somatic cell lineages were described. ScRNA-seq regularly showed major modifications around 11 years of age to eventually reach the adult state. Different spermatogonial stem cell (SSC) substates were described and scRNA-seq analyses are in favor of a paradigm shift, as the Adark and Apale spermatogonia populations could not distinctly be identified among the different SSC states. Data on the somatic cell lineage are limited, especially for Sertoli cells due technical issues related to cell size. During cell culture, scRNA-seq data showed that undifferentiated SSCs were favored in the presence of an AKT-signaling pathway inhibitor. The involvement of the oxidative phosphorylation pathway depended on the maturational state of the cells. Commonly identified cell signaling pathways during the testis development and maturation highlight factors that can be essential during specific maturation stages in IVM.


Subject(s)
Spermatogenesis , Testis , Transcriptome , Humans , Spermatogenesis/genetics , Male , Testis/metabolism , Testis/growth & development , Gene Expression Profiling/methods , Spermatogonia/metabolism , Spermatogonia/cytology , Single-Cell Analysis/methods
4.
Int J Mol Sci ; 25(13)2024 Jul 08.
Article in English | MEDLINE | ID: mdl-39000597

ABSTRACT

Drosophila spermatogenesis involves the renewal of germline stem cells, meiosis of spermatocytes, and morphological transformation of spermatids into mature sperm. We previously demonstrated that Ocnus (ocn) plays an essential role in spermatogenesis. The ValRS-m (Valyl-tRNA synthetase, mitochondrial) gene was down-regulated in ocn RNAi testes. Here, we found that ValRS-m-knockdown induced complete sterility in male flies. The depletion of ValRS-m blocked mitochondrial behavior and ATP synthesis, thus inhibiting the transition from spermatogonia to spermatocytes, and eventually, inducing the accumulation of spermatogonia during spermatogenesis. To understand the intrinsic reason for this, we further conducted transcriptome-sequencing analysis for control and ValRS-m-knockdown testes. The differentially expressed genes (DEGs) between these two groups were selected with a fold change of ≥2 or ≤1/2. Compared with the control group, 4725 genes were down-regulated (dDEGs) and 2985 genes were up-regulated (uDEGs) in the ValRS-m RNAi group. The dDEGs were mainly concentrated in the glycolytic pathway and pyruvate metabolic pathway, and the uDEGs were primarily related to ribosomal biogenesis. A total of 28 DEGs associated with mitochondria and 6 meiosis-related genes were verified to be suppressed when ValRS-m was deficient. Overall, these results suggest that ValRS-m plays a wide and vital role in mitochondrial behavior and spermatogonia differentiation in Drosophila.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Infertility, Male , Spermatogenesis , Animals , Male , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Infertility, Male/genetics , Infertility, Male/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila Proteins/deficiency , Spermatogenesis/genetics , Mitochondria/metabolism , Mitochondria/genetics , Testis/metabolism , Meiosis/genetics , Spermatogonia/metabolism , Gene Expression Profiling , Amino Acyl-tRNA Synthetases/genetics , Amino Acyl-tRNA Synthetases/metabolism , Spermatocytes/metabolism , Transcriptome
5.
Nat Commun ; 15(1): 5582, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38961093

ABSTRACT

Male infertility is a major public health concern globally with unknown etiology in approximately half of cases. The decline in total sperm count over the past four decades and the parallel increase in childhood obesity may suggest an association between these two conditions. Here, we review the molecular mechanisms through which obesity during childhood and adolescence may impair future testicular function. Several mechanisms occurring in obesity can interfere with the delicate metabolic processes taking place at the testicular level during childhood and adolescence, providing the molecular substrate to hypothesize a causal relationship between childhood obesity and the risk of low sperm counts in adulthood.


Subject(s)
Sertoli Cells , Spermatogonia , Male , Humans , Sertoli Cells/metabolism , Child , Adolescent , Spermatogonia/metabolism , Infertility, Male/metabolism , Metabolic Diseases/metabolism , Spermatogenesis , Pediatric Obesity/metabolism , Testis/metabolism , Testis/growth & development , Animals , Sperm Count
6.
Genome Biol ; 25(1): 193, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39030605

ABSTRACT

BACKGROUND: The mitosis-to-meiosis switch during spermatogenesis requires dynamic changes in gene expression. However, the regulation of meiotic transcriptional and post-transcriptional machinery during this transition remains elusive. RESULTS: We report that methyltransferase-like protein 16 (METTL16), an N6-methyladenosine (m6A) writer, is required for mitosis-to-meiosis transition during spermatogenesis. Germline conditional knockout of Mettl16 in male mice impairs spermatogonial differentiation and meiosis initiation. Mechanistically, METTL16 interacts with splicing factors to regulate the alternative splicing of meiosis-related genes such as Stag3. Ribosome profiling reveals that the translation efficiency of many meiotic genes is dysregulated in METTL16-deficient testes. m6A-sequencing shows that ablation of METTL16 causes upregulation of the m6A-enriched transcripts and downregulation of the m6A-depleted transcripts, similar to Meioc and/or Ythdc2 mutants. Further in vivo and in vitro experiments demonstrate that the methyltransferase activity site (PP185-186AA) of METTL16 is necessary for spermatogenesis. CONCLUSIONS: Our findings support a molecular model wherein the m6A writer METTL16-mediated alternative splicing and translation efficiency regulation are required to control the mitosis-to-meiosis germ cell fate decision in mice, with implications for understanding meiosis-related male fertility disorders.


Subject(s)
Adenosine , Alternative Splicing , Meiosis , Methyltransferases , Spermatogenesis , Animals , Spermatogenesis/genetics , Male , Methyltransferases/metabolism , Methyltransferases/genetics , Mice , Adenosine/analogs & derivatives , Adenosine/metabolism , Protein Biosynthesis , Mice, Knockout , Mitosis , Testis/metabolism , Spermatogonia/metabolism
7.
Virulence ; 15(1): 2384564, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39072452

ABSTRACT

Porcine reproductive and respiratory syndrome virus (PRRSV) causes a highly contagious disease that threatens the global swine industry. Recent studies have focused on the damage that PRRSV causes to the reproductive system of male pigs, although pathological research is lacking. Therefore, we examined the pathogenic mechanisms in male piglets infected with PRRSV. Gross and histopathological changes indicated that PRRSV affected the entire reproductive system, as confirmed via immunohistochemical analysis. PRRSV infected Sertoli cells and spermatogonia. To test the new hypothesis that PRRSV infection in piglets impairs blood - testis barrier (BTB) development, we investigated the pathology of PRRSV damage in the BTB. PRRSV infection significantly decreased the quantity and proliferative capacity of Sertoli cells constituting the BTB. Zonula occludens-1 and ß-catenin were downregulated in cell - cell junctions. Transcriptome analysis revealed that several crucial genes and signalling pathways involved in the growth and development of Leydig cells, Sertoli cells, and tight junctions in the testes were downregulated. Apoptosis, necroptosis, inflammatory, and oxidative stress-related pathways were activated, whereas hormone secretion-related pathways were inhibited. Many Sertoli cells and spermatogonia underwent apoptosis during early differentiation. Infected piglets exhibited disrupted androgen secretion, leading to significantly reduced testosterone and anti-Müllerian hormone levels. A cytokine storm occurred, notably upregulating cytokines such as tumour necrosis factor-α and interleukin-6. Markers of oxidative-stress damage (i.e. H2O2, malondialdehyde, and glutathione) were upregulated, whereas antioxidant-enzyme activities (i.e. superoxide dismutase, total antioxidant capacity, and catalase) were downregulated. Our results demonstrated that PRRSV infected multiple organs in the male reproductive system, which impaired growth in the BTB.


Subject(s)
Blood-Testis Barrier , Porcine Reproductive and Respiratory Syndrome , Porcine respiratory and reproductive syndrome virus , Sertoli Cells , Testis , Animals , Male , Swine , Porcine respiratory and reproductive syndrome virus/pathogenicity , Porcine respiratory and reproductive syndrome virus/physiology , Porcine Reproductive and Respiratory Syndrome/virology , Porcine Reproductive and Respiratory Syndrome/pathology , Sertoli Cells/virology , Sertoli Cells/metabolism , Blood-Testis Barrier/virology , Testis/virology , Testis/pathology , Spermatogonia/virology , Apoptosis , Leydig Cells/virology , Cytokines/metabolism , Testosterone/blood , Zonula Occludens-1 Protein/metabolism , Zonula Occludens-1 Protein/genetics
8.
Development ; 151(13)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38953252

ABSTRACT

Spermatogonial stem cell (SSC) self-renewal and differentiation provide foundational support for long-term, steady-state spermatogenesis in mammals. Here, we have investigated the essential role of RNA exosome associated DIS3 ribonuclease in maintaining spermatogonial homeostasis and facilitating germ cell differentiation. We have established male germ-cell Dis3 conditional knockout (cKO) mice in which the first and subsequent waves of spermatogenesis are disrupted. This leads to a Sertoli cell-only phenotype and sterility in adult male mice. Bulk RNA-seq documents that Dis3 deficiency partially abolishes RNA degradation and causes significant increases in the abundance of transcripts. This also includes pervasively transcribed PROMoter uPstream Transcripts (PROMPTs), which accumulate robustly in Dis3 cKO testes. In addition, scRNA-seq analysis indicates that Dis3 deficiency in spermatogonia significantly disrupts RNA metabolism and gene expression, and impairs early germline cell development. Overall, we document that exosome-associated DIS3 ribonuclease plays crucial roles in maintaining early male germ cell lineage in mice.


Subject(s)
Fertility , Spermatogonia , Testis , Animals , Male , Mice , Cell Differentiation , Exosome Multienzyme Ribonuclease Complex/metabolism , Exosome Multienzyme Ribonuclease Complex/genetics , Exosomes/metabolism , Fertility/genetics , Infertility, Male/genetics , Mice, Knockout , RNA Stability/genetics , Sertoli Cells/metabolism , Spermatogenesis , Spermatogonia/metabolism , Spermatogonia/cytology , Testis/metabolism
9.
Sci Rep ; 14(1): 15732, 2024 07 08.
Article in English | MEDLINE | ID: mdl-38977826

ABSTRACT

YAP plays a vital role in controlling growth and differentiation in various cell lineages. Although the expression of YAP in mice testicular and spermatogenic cells suggests its role in mammalian spermatogenesis, the role of YAP in the development of human male germ cells has not yet been determined. Using an in vitro model and a gene editing approach, we generated human spermatogonia stem cell-like cells (hSSLCs) from human embryonic stem cells (hESCs) and investigated the role of YAP in human spermatogenesis. The results showed that reducing YAP expression during the early stage of spermatogenic differentiation increased the number of PLZF+ hSSLCs and haploid spermatid-like cells. We also demonstrated that the up-regulation of YAP is essential for maintaining spermatogenic cell survival during the later stages of spermatogenic differentiation. The expression of YAP that deviates from this pattern results in a lower number of hSSLCs and an increased level of spermatogenic cell death. Taken together, our result demonstrates that the dynamic expression pattern of YAP is essential for human spermatogenesis. Modulating the level of YAP during human spermatogenesis could improve the production yield of male germ cells derived from hESCs, which could provide the optimization method for in vitro gametogenesis and gain insight into the application in the treatment of male infertility.


Subject(s)
Adaptor Proteins, Signal Transducing , Cell Differentiation , Human Embryonic Stem Cells , Spermatogenesis , Transcription Factors , YAP-Signaling Proteins , Male , Humans , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , YAP-Signaling Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Spermatogonia/metabolism , Spermatogonia/cytology , Promyelocytic Leukemia Zinc Finger Protein/metabolism , Promyelocytic Leukemia Zinc Finger Protein/genetics
10.
Nature ; 632(8023): 201-208, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39020172

ABSTRACT

Telomerase is intimately associated with stem cells and cancer, because it catalytically elongates telomeres-nucleoprotein caps that protect chromosome ends1. Overexpression of telomerase reverse transcriptase (TERT) enhances the proliferation of cells in a telomere-independent manner2-8, but so far, loss-of-function studies have provided no evidence that TERT has a direct role in stem cell function. In many tissues, homeostasis is shaped by stem cell competition, a process in which stem cells compete on the basis of inherent fitness. Here we show that conditional deletion of Tert in the spermatogonial stem cell (SSC)-containing population in mice markedly impairs competitive clone formation. Using lineage tracing from the Tert locus, we find that TERT-expressing SSCs yield long-lived clones, but that clonal inactivation of TERT promotes stem cell differentiation and a genome-wide reduction in open chromatin. This role for TERT in competitive clone formation occurs independently of both its reverse transcriptase activity and the canonical telomerase complex. Inactivation of TERT causes reduced activity of the MYC oncogene, and transgenic expression of MYC in the TERT-deleted pool of SSCs efficiently rescues clone formation. Together, these data reveal a catalytic-activity-independent requirement for TERT in enhancing stem cell competition, uncover a genetic connection between TERT and MYC and suggest that a selective advantage for stem cells with high levels of TERT contributes to telomere elongation in the male germline during homeostasis and ageing.


Subject(s)
Cell Competition , Clone Cells , Stem Cells , Telomerase , Animals , Male , Mice , Cell Differentiation , Cell Lineage , Chromatin/metabolism , Chromatin/genetics , Clone Cells/cytology , Clone Cells/enzymology , Clone Cells/metabolism , Gene Deletion , Genes, myc , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , Spermatogonia/cytology , Spermatogonia/metabolism , Stem Cells/cytology , Stem Cells/enzymology , Stem Cells/metabolism , Telomerase/deficiency , Telomerase/genetics , Telomerase/metabolism , Reverse Transcription , Biocatalysis , Homeostasis , Aging
11.
Cell Death Dis ; 15(7): 499, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38997255

ABSTRACT

Dynein complexes are large, multi-unit assemblies involved in many biological processes via their critical roles in protein transport and axoneme motility. Using next-generation sequencing of infertile men presenting with low or no sperm in their ejaculates, we identified damaging variants in the dynein-related gene AXDND1. We thus hypothesised that AXDND1 is a critical regulator of male fertility. To test this hypothesis, we produced a knockout mouse model. Axdnd1-/- males were sterile at all ages but presented with an evolving testis phenotype wherein they could undergo one round of histologically replete spermatogenesis followed by a rapid depletion of the seminiferous epithelium. Marker experiments identified a role for AXDND1 in maintaining the balance between differentiation-committed and self-renewing spermatogonial populations, resulting in disproportionate production of differentiating cells in the absence of AXDND1 and increased sperm production during initial spermatogenic waves. Moreover, long-term spermatogonial maintenance in the Axdnd1 knockout was compromised, ultimately leading to catastrophic germ cell loss, destruction of blood-testis barrier integrity and immune cell infiltration. In addition, sperm produced during the first wave of spermatogenesis were immotile due to abnormal axoneme structure, including the presence of ectopic vesicles and abnormalities in outer dense fibres and microtubule doublet structures. Sperm output was additionally compromised by a severe spermiation defect and abnormal sperm individualisation. Collectively these data identify AXDND1 as an atypical dynein complex-related protein with a role in protein/vesicle transport of relevance to spermatogonial function and sperm tail formation in mice and humans. This study underscores the importance of studying the consequences of gene loss-of-function on both the establishment and maintenance of male fertility.


Subject(s)
Mice, Knockout , Sperm Tail , Spermatogenesis , Spermatogonia , Animals , Humans , Male , Mice , Cell Differentiation , Dyneins/metabolism , Infertility, Male/genetics , Infertility, Male/metabolism , Infertility, Male/pathology , Mice, Inbred C57BL , Sperm Tail/metabolism , Spermatogenesis/genetics , Spermatogonia/metabolism , Testis/metabolism , Axonemal Dyneins/genetics , Axonemal Dyneins/metabolism
12.
Reprod Fertil Dev ; 362024 Jul.
Article in English | MEDLINE | ID: mdl-38991104

ABSTRACT

Context A population of sperm progenitor cells, known as Asingle spermatogonia, has been described in mammalian testes. During division cycles in spermatogenesis, some cells will form part of the Asingle spermatogonia group, while others form primary spermatocytes. Thus, during spermatogenesis, spermatogonia are the progenitor cells of spermatozoa. Aims In this study, we characterise the spermatogonial stem cells (SSCs) in the testicles of Artibeus jamaicensis and Sturnira lilium bats. The knowledge generated from this will contribute to the understanding of the biology of germ cells and the mechanisms of spermatogenesis in mammals, generating information on wildlife species that are important for biodiversity. Methods Testes were analysed by light and electron microscopy. Likewise, the expression of specific factors of stem cells (Oct4 and C-kit), germ cells (Vasa), cell proliferation (pH3 and SCP1) and testicular somatic cells (MIS, 3ßHSD and Sox9) was characterised by immunofluorescence and western blot. Key results The histological analysis enabled the location of type Asingle, Apaired and Aaligned spermatogonia in the periphery of the seminiferous tubules adjacent to Sertoli cells. The expression of genes of stem and germ cells made it possible to corroborate the distribution of the SSCs. Conclusions Results indicate that type Asingle spermatogonia were not randomly distributed, since proliferative activity was detected in groups of cells adjacent to the seminiferous tubules membrane, suggesting the localisation of spermatogonial niches in a specific region of testes. Implications This study provides evidence for the existence of SSCs in the testis of chiropterans that contribute to the renewal of germline progenitor cells to maintain the reproduction of the organisms.


Subject(s)
Chiroptera , Spermatogenesis , Spermatogonia , Testis , Animals , Male , Testis/cytology , Testis/metabolism , Spermatogonia/cytology , Spermatogenesis/physiology , Stem Cells/cytology , Cell Proliferation , Adult Germline Stem Cells/metabolism , Adult Germline Stem Cells/cytology
13.
Reprod Domest Anim ; 59(7): e14661, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38979950

ABSTRACT

Spermatogonial stem cells (SSCs) comprise the foundation of spermatogenesis and hence have great potential for fertility preservation of rare or endangered species and the development of transgenic animals and birds. Yet, developing optimal conditions for the isolation, culture, and maintenance of SSCs in vitro remains challenging, especially for chicken. The objectives of this study were to (1) find the optimal age for SSC isolation in Huaixiang chicken, (2) develop efficient protocols for the isolation, (3) enrichment, and (4) culture of isolated SSCs. In the present study, we first compared the efficiency of SSC isolation using 11 different age groups (8-79 days of age) of Huaixiang chicken. We found that the testes of 21-day-old chicken yielded the highest cell viability. Next, we compared two different enzymatic combinations for isolating SSCs and found that 0.125% trypsin and 0.02 g/L EDTA supported the highest number and viability of SSCs. This was followed by investigating optimal conditions for the enrichment of SSCs, where we observed that differential plating had the highest enrichment efficiency compared to the Percoll gradient and magnetic-activated cell sorting methods. Lastly, to find the optimal culture conditions of SSCs, we compared adding different concentrations of foetal bovine serum (FBS; 2%, 5%, 7%, and 10%) and different concentrations of GDNF, bFGF, or LIF (5, 10, 20, or 30 ng/mL). We found that a combination of 2% FBS and individual growth factors, including GDNF (20 ng/mL), bFGF (30 ng/mL), or LIF (5 ng/mL), best supported the proliferation and colony formation of SSCs. In conclusion, SSCs can be optimally isolated through enzymatic digestion from testes of 21-day-old chicken, followed by enrichment using differential plating. Furthermore, adding 2% FBS and optimized concentrations of GFNF, bFGF, or LIF in the culture promotes the proliferation of chicken SSCs.


Subject(s)
Adult Germline Stem Cells , Cell Culture Techniques , Cell Separation , Chickens , Animals , Male , Cell Culture Techniques/veterinary , Cell Separation/methods , Cell Separation/veterinary , Testis/cytology , Spermatogonia/cytology , Cell Survival , Cells, Cultured
14.
Zhonghua Nan Ke Xue ; 30(1): 9-17, 2024 Jan.
Article in Chinese | MEDLINE | ID: mdl-39046408

ABSTRACT

OBJECTIVE: To investigate the expression pattern of the D930020B18Rik gene in the testis of the mouse in different stages of development and its possible role in spermatogenesis. METHODS: Using gene expression profile microarray, we identified highly expressed D930020B18Rik in the mouse testis and analyzed the expression pattern of the gene by qPCR, immunohistochemistry, Western blot and immunofluorescence staining, and verified its function and molecular mechanism using bioinformatics analysis, dual-luciferase reporter assay and cell cycle synchronization. RESULTS: The expression of the D930020B18Rik gene remained low in the testis of the mouse and mainly localized in the cytoplasm of spermatogonia during the first 2 postnatal weeks (PNW), increased from the 3rd PNW to sexual maturity, localized in the cytoplasm of spermatogonia and the nuclei of round and elongated spermatids, but was absent in the nuclei of mature sperm. Phylogenetic analysis showed that the D930020B18Rik protein sequence was highly conserved in mammals. Gene set enrichment analysis indicated that D930020B18Rik and its homologous protein might be involved in regulating spermatogenesis of mammals by participating in nucleoplasmic condensation (normalized enrichment score ï¼»NESï¼½ = 1.652, P < 0.01, false discovery rate ï¼»FDRï¼½ = 0.153), meiosis (NES = 1.960, P < 0.01, FDR = 0.001) and formation of microtubule cytoskeleton during mitosis (NES = 1.903, P < 0.01, FDR = 0.009). Dual-luciferase reporter assay revealed that the transcription factors klf5 and foxo1 could identify and bind D930020B18Rik promoters and perform the function of positive or negative transcriptional regulation. CONCLUSION: The D930020B18Rik gene is expressed in the mouse testis in a time- and location-specific manner, highly associated with spermiogenesis, mainly localized in the nuclei of germ cells, and may be involved in the meiosis of spermatocytes and spermiogenesis.


Subject(s)
Spermatogenesis , Testis , Animals , Male , Spermatogenesis/genetics , Mice , Testis/metabolism , Spermatogonia/metabolism , Spermatogonia/cytology , Phylogeny , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Gene Expression Profiling
15.
Toxicology ; 507: 153888, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39019315

ABSTRACT

Titanium dioxide nanoparticles (TiO2 NPs) can reduce sperm number, but the mechanisms of defective spermatogenesis induced by TiO2 NPs have not been studied through cell-cell interactions at present. A kind of biomimetic three-dimensional blood-testis barrier microfluidic chip capable of intercellular communication was constructed with soft lithography techniques, including Sertoli cell (TM4), spermatogonia (GC-1) and vascular endothelial cell units, to study the mechanisms of TiO2 NPs-induced defective spermatogenesis. TM4 and GC-1 cells cultured in TiO2 NPs exposure and control chips were collected for transcriptomics and metabonomics analysis, and key proteins and metabolites in changed biological processes were validated. In TM4 cells, TiO2 NPs suppressed glucose metabolism, especially lactate production, which reduced energy substrate supply for spermatogenesis. TiO2 NPs also decreased the levels of key proteins and metabolites of lactate production. In GC-1 cells, TiO2 NPs disturbed chemokine signaling pathways regulating cell proliferation and interfered with glutathione metabolism. The Cxcl13, Stat3 and p-Stat3 levels and cell proliferation rate were decreased, and the GSR, GPX4 and GSH contents were increased in GC-1 cells in chips under TiO2 NPs treatment. The decrease in energy substrate supply for spermatogenesis and inhibition of spermatogonia proliferation could be the main mechanisms of defective spermatogenesis induced by TiO2 NPs.


Subject(s)
Blood-Testis Barrier , Sertoli Cells , Spermatogenesis , Spermatogonia , Titanium , Male , Titanium/toxicity , Spermatogenesis/drug effects , Animals , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Blood-Testis Barrier/drug effects , Mice , Spermatogonia/drug effects , Spermatogonia/metabolism , Spermatogonia/pathology , Cell Line , Metal Nanoparticles/toxicity , Lab-On-A-Chip Devices , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Cell Communication/drug effects
16.
Ecotoxicol Environ Saf ; 280: 116562, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38850704

ABSTRACT

Diquat dibromide (DQ) is a globally used herbicide in agriculture, and its overuse poses an important public health issue, including male reproductive toxicity in mammals. However, the effects and molecular mechanisms of DQ on testes are limited. In vivo experiments, mice were intraperitoneally injected with 8 or 10 mg/kg/ day of DQ for 28 days. It has been found that heme oxygenase-1 (HO-1) mediates DQ-induced ferroptosis in mouse spermatogonia, thereby damaging testicular development and spermatogenesis. Histopathologically, we found that DQ exposure caused seminiferous tubule disorders, reduced germ cells, and increased sperm malformation, in mice. Reactive oxygen species (ROS) staining of frozen section and transmission electron microscopy (TEM) displayed DQ promoted ROS generation and mitochondrial morphology alterations in mouse testes, suggesting that DQ treatment induced testicular oxidative stress. Subsequent RNA-sequencing further showed that DQ treatment might trigger ferroptosis pathway, attributed to disturbed glutathione metabolism and iron homeostasis in spermatogonia cells in vitro. Consistently, results of western blotting, measurements of MDA and ferrous iron, and ROS staining confirmed that DQ increased oxidative stress and lipid peroxidation, and accelerated ferrous iron accumulation both in vitro and in vivo. Moreover, inhibition of ferroptosis by deferoxamine (DFO) markedly ameliorated DQ-induced cell death and dysfunction. By RNA-sequencing, we found that the expression of HO-1 was significantly upregulated in DQ-treated spermatogonia, while ZnPP (a specific inhibitor of HO-1) blocked spermatogonia ferroptosis by balancing intracellular iron homeostasis. In mice, administration of the ferroptosis inhibitor ferrostatin-1 effectively restored the increase of HO-1 levels in the spermatogonia, prevented spermatogonia death, and alleviated the spermatogenesis disorders induced by DQ. Overall, these findings suggest that HO-1 mediates DQ-induced spermatogonia ferroptosis in mouse testes, and targeting HO-1 may be an effective protective strategy against male reproductive disorders induced by pesticides in agriculture.


Subject(s)
Diquat , Ferroptosis , Heme Oxygenase-1 , Herbicides , Reactive Oxygen Species , Spermatogonia , Testis , Animals , Male , Ferroptosis/drug effects , Mice , Spermatogonia/drug effects , Spermatogonia/pathology , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Testis/drug effects , Testis/pathology , Diquat/toxicity , Herbicides/toxicity , Reactive Oxygen Species/metabolism , Oxidative Stress/drug effects , Spermatogenesis/drug effects , Membrane Proteins
17.
PLoS One ; 19(6): e0304475, 2024.
Article in English | MEDLINE | ID: mdl-38848382

ABSTRACT

Cystic spermatogenesis in the subadult, maturing and adult Greenland shark (Somniosus microcephalus) displays multiple novel features, characterized early on by an unorganized internal cellular environment of the spermatocysts (anatomically discrete follicle-like units containing a single germ cell stage and its complement of co-developing Sertoli cells). These typically show polar asymmetries due to asymmetrically distributed germ and Sertoli cells. These arise from several novel cellular rearrangements at the immature pole, including fusion of a cluster of somatic cells with newly formed cysts containing only one to three spermatogonia and that already display an excess of Sertoli cells. The subadult's germinative zone revealed an additional novelty, namely numerous previously formed somatic cell-lined rings into which spermatogonia were incorporated. A striking finding was the conspicuous rarity of the routinely discernible Sertoli mitotic figures in the hallmark cyst stage of diametric elasmobranch spermatogenesis that is known for the peak display of the latter. Scrutiny of sequentially unfolding phenomena in the linearly arranged spermatogonial generations revealed that the cellular developments at the most common type of cyst-duct transition area (comprising slender to spindle-like basophilic cells with pointed ends) were concurrent with the discreet appearance of a second dark Sertoli nucleus, a development that persisted in spermiated cysts. Spermatogenically active mature males displayed vigorous meiotic divisions. However, a scattering of their spermatid cysts also displayed shark-atypical asynchronous passage through spermiogenesis, phenomena which were exacerbated as arrested spermiogenesis in an archival collection of tissues from 13 maturing specimens. Subadult specimens revealed meiotic arrest, and foci of infiltration of leukocytes that originate from a mass of eosinophilic, granule-laden immune cells dorsally under the testis capsule. This tissue was identical to the testis-affixed bone marrow equivalent in other shark species. This tissue is likely developmentally regulated in the Greenland shark as it is absent in adults.


Subject(s)
Sertoli Cells , Sharks , Spermatogenesis , Animals , Male , Sharks/physiology , Sertoli Cells/cytology , Sertoli Cells/physiology , Spermatogenesis/physiology , Spermatogonia/cytology , Testis/cytology
18.
J Insect Physiol ; 156: 104668, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38942138

ABSTRACT

During reproduction, females may boost their fitness by being selective based on direct material benefits provided by the males, such as nuptial gifts. In Lepidoptera, male provides a spermatophore containing nutrients. However, virgin males produce a bigger spermatophore, containing spermatozoa and nutrients, allowing higher female fertility. Lepidoptera females that could detect the sexual status of males may thus prefer a male without previous mating experience (i.e. a virgin male). This mate selection could be achieved by the use of chemical indices, such as sexual pheromones and cuticular compounds, known to be possibly exchanged during reproduction, and which can be indicators of a previous mating experience and known to be possibly sources of information exchanged. In this study, we experimentally presented Lobesia botrana virgin males with females in order for them to be exposed to females' natural sexual pheromones or cuticular compounds. 12 or 48 h after the exposure of males to either females' sexual pheromones or cuticular compounds, these males were confronted to naïve females, which have a choice between them or a virgin non-exposed males. We highlighted that, despite producing a spermatophore of similar volume, all exposed virgin males were less likely to mate with females 12 h after exposure, while after 48 h of exposure this is only the case for virgin males exposed to sexual pheromones. L. botrana females may thus discriminate male sexual experience based on chemical cues (either from cues transferred directly from females to males, or from changes in the cuticular or pheromone males' profile) indicating past mating experiences. Mating duration was longer for males exposed to sexual pheromones after 12 h only, and for males exposed to cuticular compounds after 48 h only. Pheromones signal might be more persistent over time and seems to more easily gather information for males. The physiological reasoning behind this result still needs to be investigated.


Subject(s)
Mating Preference, Animal , Moths , Sex Attractants , Animals , Male , Female , Moths/physiology , Sex Attractants/pharmacology , Sexual Behavior, Animal , Spermatogonia/physiology , Lobesia botrana
19.
Ecotoxicol Environ Saf ; 280: 116544, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38838463

ABSTRACT

Benzyl butyl phthalate (BBP) is a widely used plasticizer that poses various potential health hazards. Although BBP has been extensively studied, the direct mechanism underlying its toxicity in male germ cells remains unclear. Therefore, we investigated BBP-mediated male germ cell toxicity in GC-1 spermatogonia (spg), a differentiated mouse male germ cell line. This study investigated the impact of BBP on reactive oxygen species (ROS) generation, apoptosis, and autophagy regulation, as well as potential protective measures against BBP-induced toxicity. A marked dose-dependent decrease in GC-1 spg cell proliferation was observed following treatment with BBP at 12.5 µM. Exposure to 50 µM BBP, approximating the IC50 of 53.9 µM, markedly increased cellular ROS generation and instigated apoptosis, as evidenced by augmented protein levels of both intrinsic and extrinsic apoptosis-related markers. An amount of 50 µM BBP induced marked upregulation of autophagy regulator proteins, p38 MAPK, and extracellular signal-regulated kinase and substantially downregulated the phosphorylation of key kinases involved in regulating cell proliferation, including phosphoinositide 3-kinase, protein kinase B, mammalian target of rapamycin (mTOR), c-Jun N-terminal kinase. The triple combination of N-acetylcysteine, parthenolide, and 3-methyladenine markedly restored cell proliferation, decreased BBP-induced apoptosis and autophagy, and restored mTOR phosphorylation. This study provides new insights into BBP-induced male germ cell toxicity and highlights the therapeutic potential of the triple inhibitors in mitigating BBP toxicity.


Subject(s)
Acetylcysteine , Adenine , Apoptosis , Autophagy , Cell Proliferation , Phthalic Acids , Reactive Oxygen Species , Sesquiterpenes , Male , Animals , Mice , Phthalic Acids/toxicity , Autophagy/drug effects , Apoptosis/drug effects , Reactive Oxygen Species/metabolism , Sesquiterpenes/pharmacology , Acetylcysteine/pharmacology , Adenine/analogs & derivatives , Adenine/pharmacology , Adenine/toxicity , Cell Proliferation/drug effects , Cell Line , Plasticizers/toxicity , Spermatogonia/drug effects
20.
Biomolecules ; 14(6)2024 May 21.
Article in English | MEDLINE | ID: mdl-38927011

ABSTRACT

Normal testicular development ensures the process of spermatogenesis, which is a complex biological process. The sustained high productivity of spermatogenesis throughout life is predominantly attributable to the constant proliferation and differentiation of spermatogonial stem cells (SSCs). The self-renewal and differentiation processes of SSCs are strictly regulated by the SSC niche. Therefore, understanding the developmental pattern of SSCs is crucial for spermatogenesis. The Shaziling pig is a medium-sized indigenous pig breed originating from central China. It is renowned for its superior meat quality and early male sexual maturity. The spermatogenic ability of the boars is of great economic importance to the pig industry. To investigate testicular development, particularly the pattern of SSC development in Shaziling pigs, we used single-cell transcriptomics to identify gene expression patterns in 82,027 individual cells from nine Shaziling pig testes at three key postnatal developmental stages. We generated an unbiased cell developmental atlas of Shaziling pig testicular tissues. We elucidated the complex processes involved in the development of SSCs within their niche in the Shaziling pig. Specifically, we identified potential marker genes and cellular signaling pathways that regulate SSC self-renewal and maintenance. Additionally, we proposed potential novel marker genes for SSCs that could be used for SSC isolation and sorting in Shaziling pigs. Furthermore, by immunofluorescence staining of testicular tissues of different developmental ages using marker proteins (UCHL1 and KIT), the developmental pattern of the spermatogonia of Shaziling pigs was intensively studied. Our research enhances the comprehension of the development of SSCs and provides a valuable reference for breeding Shaziling pigs.


Subject(s)
RNA-Seq , Spermatogonia , Testis , Animals , Male , Swine/genetics , Spermatogonia/metabolism , Spermatogonia/cytology , Testis/metabolism , Testis/cytology , Testis/growth & development , Adult Germline Stem Cells/metabolism , Adult Germline Stem Cells/cytology , Single-Cell Analysis , Cell Differentiation/genetics , Spermatogenesis/genetics , Stem Cells/metabolism , Stem Cells/cytology , Transcriptome/genetics
SELECTION OF CITATIONS
SEARCH DETAIL