Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 150
Filtrar
1.
Development ; 151(12)2024 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-38934417

RESUMO

Spermatogonial stem cells (SSCs) undergo self-renewal division to sustain spermatogenesis. Although it is possible to derive SSC cultures in most mouse strains, SSCs from a 129 background never proliferate under the same culture conditions, suggesting they have distinct self-renewal requirements. Here, we established long-term culture conditions for SSCs from mice of the 129 background (129 mice). An analysis of 129 testes showed significant reduction of GDNF and CXCL12, whereas FGF2, INHBA and INHBB were higher than in testes of C57BL/6 mice. An analysis of undifferentiated spermatogonia in 129 mice showed higher expression of Chrna4, which encodes an acetylcholine (Ach) receptor component. By supplementing medium with INHBA and Ach, SSC cultures were derived from 129 mice. Following lentivirus transduction for marking donor cells, transplanted cells re-initiated spermatogenesis in infertile mouse testes and produced transgenic offspring. These results suggest that the requirements of SSC self-renewal in mice are diverse, which has important implications for understanding self-renewal mechanisms in various animal species.


Assuntos
Camundongos Endogâmicos C57BL , Espermatogênese , Espermatogônias , Testículo , Animais , Masculino , Camundongos , Espermatogônias/citologia , Espermatogônias/metabolismo , Espermatogênese/genética , Espermatogênese/fisiologia , Testículo/metabolismo , Testículo/citologia , Autorrenovação Celular , Células-Tronco Germinativas Adultas/metabolismo , Células-Tronco Germinativas Adultas/citologia , Células Cultivadas , Receptores Nicotínicos/metabolismo , Receptores Nicotínicos/genética , Camundongos Endogâmicos , Diferenciação Celular , Proliferação de Células , Células-Tronco/citologia , Células-Tronco/metabolismo , Camundongos Transgênicos
2.
Cell Mol Life Sci ; 81(1): 211, 2024 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-38722330

RESUMO

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.


Assuntos
Apoptose , Proliferação de Células , Espermatogênese , Tioléster Hidrolases , Via de Sinalização Wnt , Humanos , Masculino , Células-Tronco Germinativas Adultas/metabolismo , Apoptose/genética , Azoospermia/metabolismo , Azoospermia/genética , Azoospermia/patologia , beta Catenina/metabolismo , beta Catenina/genética , Proliferação de Células/genética , Proteínas de Homeodomínio/metabolismo , Proteínas de Homeodomínio/genética , Espermatogênese/genética , Espermatogônias/metabolismo , Espermatogônias/citologia , Testículo/metabolismo , Testículo/citologia , Tioléster Hidrolases/genética , Tioléster Hidrolases/metabolismo , Via de Sinalização Wnt/genética
3.
EMBO J ; 39(20): e103667, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-32815560

RESUMO

In plants, aerial organs originate continuously from stem cells in the center of the shoot apical meristem. Descendants of stem cells in the subepidermal layer are progenitors of germ cells, giving rise to male and female gametes. In these cells, mutations, including insertions of transposable elements or viruses, must be avoided to preserve genome integrity across generations. To investigate the molecular characteristics of stem cells in Arabidopsis, we isolated their nuclei and analyzed stage-specific gene expression and DNA methylation in plants of different ages. Stem cell expression signatures are largely defined by developmental stage but include a core set of stem cell-specific genes, among which are genes implicated in epigenetic silencing. Transiently increased expression of transposable elements in meristems prior to flower induction correlates with increasing CHG methylation during development and decreased CHH methylation, before stem cells enter the reproductive lineage. These results suggest that epigenetic reprogramming may occur at an early stage in this lineage and could contribute to genome protection in stem cells during germline development.


Assuntos
Proteínas de Arabidopsis/metabolismo , Arabidopsis/genética , Arabidopsis/metabolismo , Metilação de DNA , Elementos de DNA Transponíveis/genética , Brotos de Planta/metabolismo , Células-Tronco/metabolismo , Células-Tronco Germinativas Adultas/metabolismo , Arabidopsis/crescimento & desenvolvimento , Proteínas de Arabidopsis/genética , Núcleo Celular/genética , Núcleo Celular/metabolismo , Epigênese Genética , Epigenômica , Perfilação da Expressão Gênica , Regulação da Expressão Gênica de Plantas/genética , Ontologia Genética , Inativação Gênica , Estudo de Associação Genômica Ampla , Meristema/genética , Meristema/crescimento & desenvolvimento , Meristema/metabolismo , Brotos de Planta/crescimento & desenvolvimento , Análise de Componente Principal , RNA-Seq
4.
Reproduction ; 168(1)2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38670156

RESUMO

In brief: Oogonial stem cells in the adult ovary can generate oocytes, but they are usually quiescent. TGFB1 is key in stimulating the proliferation of OSC, thereby ensuring the sustained reproductive potential in poultry species. Abstract: Oogonial stem cells (OSCs) are a type of germ stem cell present in the adult ovary. They have the ability to self-renew through mitosis and differentiate into oocytes through meiosis. We have previously identified a population of OSCs in the chicken ovary, but the underlying mechanisms controlling their activation and proliferation were unclear. In this study, we observed that OSCs showed robust proliferation when cultured on a layer of chicken embryo fibroblasts (CEF), suggesting that CEF may secrete certain crucial factors that activate OSC proliferation. We further detected TGFB1 as a potent signaling molecule to promote OSC proliferation. Additionally, we revealed the signaling pathways that play important roles downstream of TGFB1-induced OSC proliferation. These findings provide insights into the mechanisms underlying OSC proliferation in chickens and offer a foundation for future research on in situ activation of OSC proliferation in ovary and improvement of egg-laying performance in chickens.


Assuntos
Proliferação de Células , Galinhas , Fator de Crescimento Transformador beta1 , Animais , Fator de Crescimento Transformador beta1/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Feminino , Células Cultivadas , Embrião de Galinha , Oogônios/citologia , Oogônios/metabolismo , Oogônios/fisiologia , Ovário/citologia , Ovário/metabolismo , Transdução de Sinais , Fibroblastos/citologia , Fibroblastos/metabolismo , Células-Tronco Germinativas Adultas/citologia , Células-Tronco Germinativas Adultas/metabolismo , Células-Tronco Germinativas Adultas/fisiologia
5.
J Biol Chem ; 298(2): 101559, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34979097

RESUMO

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.


Assuntos
Células-Tronco Germinativas Adultas , Cromatina , Espermatogênese , Espermatogônias , Células-Tronco Germinativas Adultas/citologia , Células-Tronco Germinativas Adultas/metabolismo , Animais , Diferenciação Celular/fisiologia , Cromatina/metabolismo , Masculino , Espermatogênese/fisiologia , Espermatogônias/citologia , Suínos
6.
Mol Reprod Dev ; 90(5): 275-286, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36966461

RESUMO

Meiosis, a key step in spermatogenesis, is affected by many factors. Current studies have shown that long noncoding RNAs (lncRNAs) are potential factors regulating meiosis, and their regulatory mechanisms have received much attention. However, little research has been done on its regulatory mechanism in the spermatogenesis of roosters. Here, we found that lncRNA involved in meiosis and spermatogenesis (lncRNA-IMS) was involved in the regulation of Stra8 by gga-miR-31-5p and hindered the inhibition of Stra8 by gga-miR-31-5p. The acquisition and loss of function experiments demonstrated that lncRNA-IMS was involved in meiosis and spermatogenesis. In addition, we predicted and determined the core promoter region of lncRNA-IMS. Prediction of transcription factors, deletion/overexpression of binding sites, knockdown/overexpression of Jun, and dual-luciferase reporter analysis confirmed that Jun positively activated transcription of lncRNA-IMS. Our findings further enrich the TF-lncRNA-miRNA-mRNA regulatory network during male meiosis and provide new ideas for studying the molecular mechanism of meiosis and spermatogenesis in chicken spermatogonial stem cells.


Assuntos
Células-Tronco Germinativas Adultas , Proteínas Aviárias , Meiose , MicroRNAs , RNA Longo não Codificante , Animais , Masculino , Células-Tronco Germinativas Adultas/metabolismo , Galinhas/genética , Galinhas/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Proteínas Aviárias/metabolismo
7.
PLoS Biol ; 18(12): e3001003, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33315855

RESUMO

Stem-cell niche signaling is short-range in nature, such that only stem cells but not their differentiating progeny receive self-renewing signals. At the apical tip of the Drosophila testis, 8 to 10 germline stem cells (GSCs) surround the hub, a cluster of somatic cells that organize the stem-cell niche. We have previously shown that GSCs form microtubule-based nanotubes (MT-nanotubes) that project into the hub cells, serving as the platform for niche signal reception; this spatial arrangement ensures the reception of the niche signal specifically by stem cells but not by differentiating cells. The receptor Thickveins (Tkv) is expressed by GSCs and localizes to the surface of MT-nanotubes, where it receives the hub-derived ligand Decapentaplegic (Dpp). The fate of Tkv receptor after engaging in signaling on the MT-nanotubes has been unclear. Here we demonstrate that the Tkv receptor is internalized into hub cells from the MT-nanotube surface and subsequently degraded in the hub cell lysosomes. Perturbation of MT-nanotube formation and Tkv internalization from MT-nanotubes into hub cells both resulted in an overabundance of Tkv protein in GSCs and hyperactivation of a downstream signal, suggesting that the MT-nanotubes also serve a second purpose to dampen the niche signaling. Together, our results demonstrate that MT-nanotubes play dual roles to ensure the short-range nature of niche signaling by (1) providing an exclusive interface for the niche ligand-receptor interaction; and (2) limiting the amount of stem cell receptors available for niche signal reception.


Assuntos
Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Receptores de Superfície Celular/metabolismo , Receptores de Superfície Celular/fisiologia , Nicho de Células-Tronco/fisiologia , Células-Tronco/metabolismo , Células-Tronco Germinativas Adultas/metabolismo , Células-Tronco Germinativas Adultas/fisiologia , Animais , Diferenciação Celular/fisiologia , Drosophila melanogaster/metabolismo , Células Germinativas/citologia , Células Germinativas/metabolismo , Ligantes , Masculino , Microtúbulos/metabolismo , Microtúbulos/fisiologia , Transdução de Sinais/fisiologia , Células-Tronco/citologia , Testículo/metabolismo
8.
PLoS Genet ; 16(3): e1008650, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32196486

RESUMO

Stem cell systems are essential for the development and maintenance of polarized tissues. Intercellular signaling pathways control stem cell systems, where niche cells signal stem cells to maintain the stem cell fate/self-renewal and inhibit differentiation. In the C. elegans germline, GLP-1 Notch signaling specifies the stem cell fate, employing the sequence-specific DNA binding protein LAG-1 to implement the transcriptional response. We undertook a comprehensive genome-wide approach to identify transcriptional targets of GLP-1 signaling. We expected primary response target genes to be evident at the intersection of genes identified as directly bound by LAG-1, from ChIP-seq experiments, with genes identified as requiring GLP-1 signaling for RNA accumulation, from RNA-seq analysis. Furthermore, we performed a time-course transcriptomics analysis following auxin inducible degradation of LAG-1 to distinguish between genes whose RNA level was a primary or secondary response of GLP-1 signaling. Surprisingly, only lst-1 and sygl-1, the two known target genes of GLP-1 in the germline, fulfilled these criteria, indicating that these two genes are the primary response targets of GLP-1 Notch and may be the sole germline GLP-1 signaling protein-coding transcriptional targets for mediating the stem cell fate. In addition, three secondary response genes were identified based on their timing following loss of LAG-1, their lack of a LAG-1 ChIP-seq peak and that their glp-1 dependent mRNA accumulation could be explained by a requirement for lst-1 and sygl-1 activity. Moreover, our analysis also suggests that the function of the primary response genes lst-1 and sygl-1 can account for the glp-1 dependent peak protein accumulation of FBF-2, which promotes the stem cell fate and, in part, for the spatial restriction of elevated LAG-1 accumulation to the stem cell region.


Assuntos
Células-Tronco Germinativas Adultas/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Ligação a DNA/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Receptores Notch/metabolismo , Células-Tronco Germinativas Adultas/citologia , Animais , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/genética , Diferenciação Celular/fisiologia , Linhagem da Célula , Proteínas de Ligação a DNA/genética , Células Germinativas/metabolismo , Peptídeo 1 Semelhante ao Glucagon/genética , RNA/metabolismo , RNA Mensageiro/metabolismo , Receptores Notch/genética , Transdução de Sinais , Células-Tronco/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
9.
Dev Biol ; 473: 105-118, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33610541

RESUMO

Gametogenesis is one of the most extreme cellular differentiation processes that takes place in Drosophila male and female germlines. This process begins at the germline stem cell, which undergoes asymmetric cell division (ACD) to produce a self-renewed daughter that preserves its stemness and a differentiating daughter cell that undergoes epigenetic and genomic changes to eventually produce haploid gametes. Research in molecular genetics and cellular biology are beginning to take advantage of the continually advancing genomic tools to understand: (1) how germ cells are able to maintain their identity throughout the adult reproductive lifetime, and (2) undergo differentiation in a balanced manner. In this review, we focus on the epigenetic mechanisms that address these two questions through their regulation of germline-soma communication to ensure germline stem cell identity and activity.


Assuntos
Células-Tronco Germinativas Adultas/fisiologia , Diferenciação Celular/genética , Gametogênese/genética , Células-Tronco Germinativas Adultas/metabolismo , Animais , Divisão Celular Assimétrica , Drosophila/embriologia , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Epigênese Genética/genética , Epigenômica/métodos , Gametogênese/fisiologia , Expressão Gênica/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Células Germinativas/metabolismo , Células-Tronco/citologia
10.
J Cell Physiol ; 237(9): 3640-3650, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35790000

RESUMO

Detailed analysis of the regulatory mechanism of spermatogonia stem cell (SSCs) genesis can provide a novel strategy for the application of SSCs in the fields of transgenic animal production and regenerative medicine. Previous studies in this study showed that WNT signaling can positively regulate the formation of SSCs, but the exact regulatory mechanism is not clear. Here, we predicted the target gene of the Wnt/TCF7L2 pathway, namely TDRD1, by bioinformatics analysis. Functional studies revealed that overexpression of TDRD1 during RA-induced SSCs formation in vitro significantly upregulated the expression of reproductive marker genes (Integrinß1 and Integrinα6), and further flow cytometric analysis also confirmed that the formation efficiency of SSCs was significantly increased after overexpression of TDRD1; while interference with TDRD1 showed the exact opposite result. The in vivo experiments were consistent with the results of the in vitro experiments. Interestingly, although Wnt/TCF7L2 can promote the formation of SSCs, its function must be dependent on the expression of TDRD1, which was also repeatedly demonstrated as a target gene of the Wnt/TCF7L2 signaling pathway. Mechanistically, we found a large number of CpG sites in the TDRD1 promoter, and BSP analysis also confirmed that DNA methylation modifications in the TDRD1 promoter were significantly higher in embryonic stem cells than in SSCs, and further dual-luciferase reporter system assays revealed that low DNA methylation modification levels could enhance TDRD1 promoter activity; although previous studies demonstrated that TCF7L2 could enrich in the TDRD1 promoter region, the binding of the two was dependent on low DNA methylation modification. Taken together, we confirmed that low DNA methylation mediates Wnt/TCF7L2 regulation of TDRD1 to promote the formation of SSCs, providing a basis for SSCs in improving animal productivity.


Assuntos
Células-Tronco Germinativas Adultas , Via de Sinalização Wnt , Células-Tronco Germinativas Adultas/metabolismo , Animais , DNA/metabolismo , Metilação de DNA/genética , Células-Tronco Embrionárias/metabolismo , Masculino , Espermatogônias/metabolismo , Via de Sinalização Wnt/genética
11.
J Cell Physiol ; 237(9): 3565-3577, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35699595

RESUMO

Studies on gene regulation and signaling transduction pathways of human spermatogonial stem cells (SSCs) are of the utmost significance for unveiling molecular mechanisms underlying human spermatogenesis and gene therapy of male infertility. We have demonstrated, for the first time, that RNF144B stimulated cell proliferation and inhibited the apoptosis of human SSCs. The target of RNF144B was identified as FCER2 by RNA sequencing. We revealed that RNF144B interacted with FCER2 by immunoprecipitation. Consistently, overexpression of FCER2 reversed the phenotype of proliferation and apoptosis of human SSCs caused by RNF144B knockdown. Interestingly, FCER2 pulled down N2ICD (NOTCH2 intracellular domain), while N2ICD could bind to FCER2 in human SSCs. The levels of NOTCH2, FCER2, HES1, and HEY1 were reduced by RNF144B siRNA in human SSCs. Significantly, RNF144B was expressed at a lower level in nonobstructive azoospermia (NOA) patients than in the obstructive azoospermia (OA) patients with normal spermatogenesis, and 52 patients with heterozygous mutations of RNF144B were detected in 1,000 NOA patients. These results implicate that RNF144B promotes the proliferation of human SSCs and suppresses their apoptosis via the FCER2/NOTCH2/HES1 pathway and that the abnormality of RNF144B is associated with spermatogenesis failure. This study thus provides novel molecular mechanisms regulating the fate determinations of human SSCs, and it offers new biomarkers for the diagnosis and treatment of male infertility.


Assuntos
Células-Tronco Germinativas Adultas , Apoptose , Azoospermia , Infertilidade Masculina , Espermatogênese , Células-Tronco Germinativas Adultas/metabolismo , Apoptose/genética , Azoospermia/complicações , Azoospermia/genética , Proliferação de Células/genética , Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Infertilidade Masculina/etiologia , Infertilidade Masculina/genética , Infertilidade Masculina/metabolismo , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Masculino , Receptor Notch2/genética , Receptor Notch2/metabolismo , Receptores de IgE/metabolismo , Espermatogênese/genética , Fatores de Transcrição HES-1/genética , Fatores de Transcrição HES-1/metabolismo
12.
Biol Reprod ; 107(5): 1331-1344, 2022 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-35980806

RESUMO

Spermatogenesis is sustained by homeostatic balance between the self-renewal and differentiation of spermatogonial stem cells, which is dependent on the strict regulation of transcription factor and chromatin modulator gene expression. Chromodomain helicase DNA-binding protein 4 is highly expressed in spermatogonial stem cells but roles in mouse spermatogenesis are not fully understood. Here, we report that the germ-cell-specific deletion of chromodomain helicase DNA-binding protein 4 resulted in complete infertility in male mice, with rapid loss of spermatogonial stem cells and excessive germ cell apoptosis. Chromodomain helicase DNA-binding protein 4-knockdown in cultured spermatogonial stem cells also promoted the expression of apoptosis-related genes and thereby activated the tumor necrosis factor signaling pathway. Mechanistically, chromodomain helicase DNA-binding protein 4 occupies the genomic regulatory region of key apoptosis-related genes, including Jun and Nfkb1. Together, our findings reveal the determinant role of chromodomain helicase DNA-binding protein 4 in spermatogonial stem cells survival in vivo, which will offer insight into the pathogenesis of male sterility and potential novel therapeutic targets.


Assuntos
Células-Tronco Germinativas Adultas , Animais , Masculino , Camundongos , Células-Tronco Germinativas Adultas/metabolismo , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Espermatogênese/genética , Espermatogônias/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , DNA Helicases/genética , DNA Helicases/metabolismo
13.
Cell Tissue Res ; 387(1): 131-142, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34725717

RESUMO

RNA N6-methyladenosine (m6A) is essential for many bioprocesses in many species, but its role in goat testis development remains elusive, especially alkB homolog 5 (ALKBH5), one of the m6A demethylases. To this end, nine healthy Haimen goats of different ages were chosen randomly to provide testes. The results showed that the expression level of ALKBH5 was increased significantly (P < 0.05) in the 9-month group compared with the 0-day and 3-month groups, and ALKBH5 was located in goat spermatocytes with the highest expression level compared with Leydig cells and Sertoli cells. Thus, pcDNA3.1-ALKBH5 was constructed to explore the influences of the ALKBH5 increase in goat spermatogonial stem cells (SSC) in vitro. The results showed that the expression level of ALKBH5 in SSC transfected with pcDNA3.1-ALKBH5 (OE_ALKBH5) was significantly increased (P < 0.001) compared with that in SSC transfected with pcDNA3.1-EGFP (EGFP). With ALKBH5 overexpression in SSC, flow cytometry analysis showed that cells at G1 phase were significantly reduced (P < 0.01), while cells at S phase significantly increased (P < 0.01), and cell apoptosis was inhibited. Accordingly, the mRNA degradation of CCND1, CCNE1, and BCL2 was suppressed with ALKBH5 overexpression in SSC after treatment with actinomycin D. Furthermore, the mRNA levels of pluripotency maintenance- and cell differentiation-associated genes were changed between the two groups. Overall, the results indicated the crucial role of ALKBH5 during Haimen goat testis development. The results of this study provide a theoretical basis and technical means for RNA methylation participating in goat testis development.


Assuntos
Células-Tronco Germinativas Adultas/metabolismo , Enzimas AlkB/metabolismo , Espermatogônias/metabolismo , Testículo/fisiologia , Animais , Diferenciação Celular , Cabras , Humanos , Masculino , Transfecção
14.
PLoS Biol ; 17(6): e3000309, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31166944

RESUMO

During suboptimal growth conditions, Caenorhabditis elegans larvae undergo a global developmental arrest called "dauer." During this stage, the germline stem cells (GSCs) become quiescent in an AMP-activated Protein Kinase (AMPK)-dependent manner, and in the absence of AMPK, the GSCs overproliferate and lose their reproductive capacity, leading to sterility when mutant animals resume normal growth. These defects correlate with the altered abundance and distribution of a number of chromatin modifications, all of which can be corrected by disabling components of the endogenous small RNA pathway, suggesting that AMPK regulates germ cell integrity by targeting an RNA interference (RNAi)-like pathway during dauer. The expression of AMPK in somatic cells restores all the germline defects, potentially through the transmission of small RNAs. Our findings place AMPK at a pivotal position linking energy stress detected in the soma to a consequent endogenous small RNA-mediated adaptation in germline gene expression, thereby challenging the "permeability" of the Weismann barrier.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Células-Tronco Germinativas Adultas/metabolismo , Interferência de RNA/fisiologia , Proteínas Quinases Ativadas por AMP/fisiologia , Células-Tronco Germinativas Adultas/fisiologia , Animais , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Células Germinativas/metabolismo , Larva/genética , RNA/metabolismo , Células-Tronco/metabolismo
15.
PLoS Genet ; 15(7): e1008062, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31295251

RESUMO

Stem cells rely on instructive cues from their environment. Alterations in microenvironments might contribute to tissue dysfunction and disease pathogenesis. Germline stem cells (GSCs) and cyst stem cells (CySC) in Drosophila testes are normally maintained in the apical area by the testicular hub. In this study, we found that reproduction leads to accumulation of early differentiating daughters of CySCs and GSCs in the testes of aged male flies, due to hyperactivation of Jun-N-terminal kinase (JNK) signaling to maintain self-renewal gene expression in the differentiating cyst cells. JNK activity is normally required to maintain CySCs in the apical niche. A muscle sheath surrounds the Drosophila testis to maintain its long coiled structure. Importantly, reproduction triggers accumulation of the tumor necrosis factor (TNF) Eiger in the testis muscle to activate JNK signaling via the TNF receptor Grindelwald in the cyst cells. Reducing Eiger activity in the testis muscle sheath suppressed reproduction-induced differentiation defects, but had little effect on testis homeostasis of unmated males. Our results reveal that reproduction in males provokes a dramatic shift in the testicular microenvironment, which impairs tissue homeostasis and spermatogenesis in the testes.


Assuntos
Células-Tronco Germinativas Adultas/citologia , Drosophila melanogaster/fisiologia , Reprodução , Células-Tronco Germinativas Adultas/metabolismo , Animais , Diferenciação Celular , Autorrenovação Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Feminino , Homeostase , Sistema de Sinalização das MAP Quinases , Masculino , Proteínas de Membrana/metabolismo , Comportamento Sexual Animal , Espermatogênese , Nicho de Células-Tronco , Testículo/citologia , Testículo/metabolismo
16.
PLoS Genet ; 15(5): e1008139, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31050682

RESUMO

Accumulating evidence indicates that paternal age correlates with disease risk in children. De novo gain-of-function mutations in the FGF-RAS-MAPK signaling pathway are known to cause a subset of genetic diseases associated with advanced paternal age, such as Apert syndrome, achondroplasia, Noonan syndrome, and Costello syndrome. It has been hypothesized that adult spermatogonial stem cells with pathogenic mutations are clonally expanded over time and propagate the mutations to offspring. However, no model system exists to interrogate mammalian germline stem cell competition in vivo. In this study, we created a lineage tracing system, which enabled undifferentiated spermatogonia with endogenous expression of HrasG12V, a known pathogenic gain-of-function mutation in RAS-MAPK signaling, to compete with their wild-type counterparts in the mouse testis. Over a year of fate analysis, neither HrasG12V-positive germ cells nor sperm exhibited a significant expansion compared to wild-type neighbors. Short-term stem cell capacity as measured by transplantation analysis was also comparable between wild-type and mutant groups. Furthermore, although constitutively active HRAS was detectable in the mutant cell lines, they did not exhibit a proliferative advantage or an enhanced response to agonist-evoked pERK signaling. These in vivo and in vitro results suggest that mouse spermatogonial stem cells are functionally resistant to a heterozygous HrasG12V mutation in the endogenous locus and that mechanisms could exist to prevent such harmful mutations from being expanded and transmitted to the next generation.


Assuntos
Células-Tronco Germinativas Adultas/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Espermatogônias/metabolismo , Células-Tronco Germinativas Adultas/fisiologia , Animais , Mutação com Ganho de Função/genética , Mutação em Linhagem Germinativa/genética , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Mutação/genética , Idade Paterna , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/fisiologia , Seleção Genética/genética , Transdução de Sinais/genética , Espermatogônias/fisiologia , Espermatozoides/metabolismo , Testículo/metabolismo
17.
Proc Natl Acad Sci U S A ; 116(33): 16404-16409, 2019 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-31358627

RESUMO

Because spermatogonial stem cells (SSCs) are immortal by serial transplantation, SSC aging in intact testes is considered to be caused by a deteriorated microenvironment. Here, we report a cell-intrinsic mode of SSC aging by glycolysis activation. Using cultured SSCs, we found that aged SSCs proliferated more actively than young SSCs and showed enhanced glycolytic activity. Moreover, they remained euploid and exhibited stable androgenetic imprinting patterns with robust SSC activity despite having shortened telomeres. Aged SSCs showed increased Wnt7b expression, which was associated with decreased Polycomb complex 2 activity. Our results suggest that aberrant Wnt7b expression activated c-jun N-terminal kinase (JNK), which down-regulated mitochondria numbers by suppressing Ppargc1a Down-regulation of Ppargc1a probably decreased reactive oxygen species and enhanced glycolysis. Analyses of the Klotho-deficient aging mouse model and 2-y-old aged rats confirmed JNK hyperactivation and increased glycolysis. Therefore, not only microenvironment but also intrinsic activation of JNK-mediated glycolysis contributes to SSC aging.


Assuntos
Envelhecimento/genética , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Proteínas Proto-Oncogênicas/genética , Espermatogênese/genética , Proteínas Wnt/genética , Células-Tronco Germinativas Adultas/metabolismo , Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Animais , Proliferação de Células/genética , Regulação da Expressão Gênica no Desenvolvimento , Glucuronidase/genética , Glicólise/genética , Proteínas Klotho , Masculino , Camundongos , Proteínas do Grupo Polycomb/genética , Ratos , Espécies Reativas de Oxigênio/metabolismo , Espermatogônias/crescimento & desenvolvimento , Espermatogônias/metabolismo , Nicho de Células-Tronco/genética , Testículo/crescimento & desenvolvimento , Testículo/metabolismo
18.
J Cell Physiol ; 236(2): 1391-1400, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32749682

RESUMO

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.


Assuntos
Proteína Morfogenética Óssea 4/genética , Epigênese Genética/genética , Histona Metiltransferases/genética , Testículo/crescimento & desenvolvimento , Células-Tronco Germinativas Adultas/citologia , Células-Tronco Germinativas Adultas/metabolismo , Animais , Blastoderma/crescimento & desenvolvimento , Diferenciação Celular/genética , Galinhas/genética , Galinhas/crescimento & desenvolvimento , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Genitália/crescimento & desenvolvimento , Células Germinativas/crescimento & desenvolvimento , Masculino , Transdução de Sinais/genética , Testículo/metabolismo , Proteína 2 Semelhante ao Fator 7 de Transcrição/genética , Fator de Crescimento Transformador beta/genética , Proteína Wnt-5a/genética
19.
J Cell Physiol ; 236(5): 3536-3551, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-32996162

RESUMO

Orchitis is one of the leading causes of male animal infertility and is associated with inflammatory reactions caused by the bacterium. It has been reported that there is a mutual coupling effect between endoplasmic reticulum stress (ERS) and inflammatory response. Our studies showed that lipopolysaccharide (LPS) could cause testicular damages, apoptosis, ERS, and inflammatory responses in spermatogonial stem cells (SSCs); ERS-related apoptosis proteins were activated and the expression of ERS genes was significantly upregulated; meanwhile, the expression of Toll-like receptor 4 and inflammation factors was apparently increased with LPS treatment. Moreover, melatonin (MEL) could rescue testicular damage, and significantly inhibited the expression of ERS-related apoptosis genes, ERS markers, and inflammatory factors in SSCs and MEL played repairing and anti-infection roles in LPS-induced testicular damage. Therefore, MEL may be used as a drug to prevent and control bacterial infections in male reproductive systems. However, the specific molecular mechanism of MEL to resist ERS and inflammatory response remains to be further studied.


Assuntos
Células-Tronco Germinativas Adultas/patologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Inflamação/patologia , Melatonina/farmacologia , Células-Tronco Germinativas Adultas/efeitos dos fármacos , Células-Tronco Germinativas Adultas/metabolismo , Animais , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Lipopolissacarídeos , Masculino , Camundongos , Modelos Biológicos , Receptores de Melatonina/metabolismo , Testículo/efeitos dos fármacos , Testículo/patologia
20.
Biol Reprod ; 105(3): 761-766, 2021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34250539

RESUMO

More than a decade ago, the ENCODE and NIH Epigenomics Roadmap consortia organized large multilaboratory efforts to profile the epigenomes of >110 different mammalian somatic cell types. This generated valuable publicly accessible datasets that are being mined to reveal genome-wide patterns of a variety of different epigenetic parameters. This consortia approach facilitated the powerful and comprehensive multiparametric integrative analysis of the epigenomes in each cell type. However, no germ cell types were included among the cell types characterized by either of these consortia. Thus, comprehensive epigenetic profiling data are not generally available for the most evolutionarily important cells, male and female germ cells. We discuss the need for reproductive biologists to generate similar multiparametric epigenomic profiling datasets for both male and female germ cells at different developmental stages and summarize our recent effort to derive such data for mammalian spermatogonial stem cells and progenitor spermatogonia.


Assuntos
Células-Tronco Germinativas Adultas/metabolismo , Epigenoma , Epigenômica , Óvulo/crescimento & desenvolvimento , Espermatozoides/crescimento & desenvolvimento , Animais , Diferenciação Celular , Epigênese Genética , Feminino , Masculino , Mamíferos , Espermatogônias/crescimento & desenvolvimento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA