Sertoli Cell-Only Syndrome

仅支持细胞综合征
  • 文章类型: Journal Article
    精原干细胞(SSC)对于持续的精子发生和男性生育能力至关重要。小鼠SSC中选择性剪接(AS)的潜在机制仍不清楚。我们证明了SRSF1对于小鼠SSC中的基因表达和剪接至关重要。交联免疫沉淀和测序数据显示,精原细胞相关基因(例如Plzf,Id4,Setdb1,Stra8,Tial1/Tiar,Bcas2,Ddx5,Srsf10,Uhrf1和Bud31)在小鼠睾丸中被SRSF1结合。小鼠生殖细胞中Srsf1的特异性缺失会损害前体SSC的归巢,导致男性不育。整装染色数据显示成年条件敲除(cKO)小鼠睾丸中不存在生殖细胞,这表明cKO小鼠中的仅支持细胞综合征。精原细胞相关基因的表达(例如Gfra1,Pou5f1,Plzf,在cKO小鼠的睾丸中Dnd1,Stra8和Taf4b)显着降低。此外,多组学分析表明,SRSF1可能通过AS直接结合和调节Tial1/Tiar的表达来影响精原细胞的存活。此外,免疫沉淀质谱和共免疫沉淀数据显示SRSF1与RNA剪接相关蛋白(例如SART1、RBM15和SRSF10)相互作用。总的来说,我们的数据揭示了SRSF1在精原细胞存活中的关键作用,这可能提供一个框架来阐明前体SSC归巢的转录后网络的分子机制。
    Spermatogonial stem cells (SSCs) are essential for continuous spermatogenesis and male fertility. The underlying mechanisms of alternative splicing (AS) in mouse SSCs are still largely unclear. We demonstrated that SRSF1 is essential for gene expression and splicing in mouse SSCs. Crosslinking immunoprecipitation and sequencing data revealed that spermatogonia-related genes (e.g. Plzf, Id4, Setdb1, Stra8, Tial1/Tiar, Bcas2, Ddx5, Srsf10, Uhrf1, and Bud31) were bound by SRSF1 in the mouse testes. Specific deletion of Srsf1 in mouse germ cells impairs homing of precursor SSCs leading to male infertility. Whole-mount staining data showed the absence of germ cells in the testes of adult conditional knockout (cKO) mice, which indicates Sertoli cell-only syndrome in cKO mice. The expression of spermatogonia-related genes (e.g. Gfra1, Pou5f1, Plzf, Dnd1, Stra8, and Taf4b) was significantly reduced in the testes of cKO mice. Moreover, multiomics analysis suggests that SRSF1 may affect survival of spermatogonia by directly binding and regulating Tial1/Tiar expression through AS. In addition, immunoprecipitation mass spectrometry and co-immunoprecipitation data showed that SRSF1 interacts with RNA splicing-related proteins (e.g. SART1, RBM15, and SRSF10). Collectively, our data reveal the critical role of SRSF1 in spermatogonia survival, which may provide a framework to elucidate the molecular mechanisms of the posttranscriptional network underlying homing of precursor SSCs.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    高通量单细胞RNA测序(scRNA-seq)广泛用于精子发生。然而,它只显示了生殖细胞和体细胞中的短读数,限制了新转录本和基因的发现。
    目的:这项研究显示了梗阻性无精子症(OA)和仅支持细胞(SCO)患者精子发生的长阅读转录图。
    方法:从OA和NOA患者的睾丸活检组织中分离单细胞。通过比较Pacbio长读数单细胞测序(OAn=3,NOAn=3)与短读数scRNA-seq(OAn=6,NOAn=6)来鉴定细胞培养。根据已知标记对10种生殖细胞类型和8种体细胞类型进行分类。
    方法:Pacbiolong读取单细胞测序,短读scRNA-seq,聚合酶链反应。
    结果:总共130,426份长读转录本(100,517份新转录本和29,909份已知转录本)和49,508份长读转录本(26,002份新转录本,和23,506已知转录物)已在OA和NOA患者中检测到,分别。此外,在OA和NOA患者中鉴定出36,373和1642个新基因,分别。重要的是,在正常精子发生过程中,在生殖细胞和气孔细胞中检测到长阅读转录本的特异性表达。
    结论:我们已经确定了OA和NOA的全长转录本,发现了新的基因.此外,检测到特异性表达的全长转录本,转录本的基因组结构被定位在不同的细胞类型中。这些发现可能为人类精子发生和男性不育的治疗提供有价值的信息。
    BACKGROUND: High-throughput single-cell RNA sequencing (scRNA-seq) is widely used in spermatogenesis. However, it only reveals short reads in germ and somatic cells, limiting the discovery of novel transcripts and genes.
    OBJECTIVE: This study shows the long-read transcriptional landscape of spermatogenesis in obstructive azoospermia (OA) and Sertoli cell-only patients.
    METHODS: Single cells were isolated from testicular biopsies of OA and non-obstructive azoospermia (NOA) patients. Cell culture was identified by comparing PacBio long-read single-cell sequencing (OA n = 3, NOA n = 3) with short-read scRNA-seq (OA n = 6, NOA n = 6). Ten germ cell types and eight somatic cell types were classified based on known markers.
    METHODS: PacBio long-read single-cell sequencing, short-read scRNA-seq, polymerase chain reaction.
    RESULTS: A total of 130 426 long-read transcripts (100 517 novel transcripts and 29 909 known transcripts) and 49 508 long-read transcripts (26 002 novel transcripts and 23 506 known transcripts) have been detected in OA and NOA patients, respectively. Moreover, 36 373 and 1642 new genes are identified in OA and NOA patients, respectively. Importantly, specific expressions of long-read transcripts were detected in germ and stomatic cells during normal spermatogenesis.
    CONCLUSIONS: We have identified total full-length transcripts in OA and NOA, and new genes were found. Furthermore, specific expressed full-length transcripts were detected, and the genomic structure of transcripts was mapped in different cell types. These findings may provide valuable information on human spermatogenesis and the treatment of male infertility.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    仅支持细胞综合征(SCOS),严重的睾丸生精失败,其特征在于完全不存在雄性生殖细胞。为了更好地扩展对SCOS潜在分子机制的理解,我们使用来自基因表达Omnibus(GEO)和ArrayExpress数据库的微阵列数据集来确定差异表达基因(DEGs)。此外,进行了功能富集分析,包括基因本体论(GO)和京都基因和基因组百科全书(KEGG)。蛋白质-蛋白质相互作用(PPI)网络,模块,和miRNA-mRNA调控网络的构建和分析,并对hub基因进行验证。总共确定了601个共享DEG,包括416个下调基因和185个上调基因。富集分析的结果表明,共享的DEG大多在有性生殖中富集,生殖过程,雄配子一代,免疫反应,和免疫相关途径。此外,六个hub基因(CCNA2、CCNB2、TOP2A、通过使用cytoHubba和MCODE插件从PPI网络中选择CDC20,BUB1和BUB1B)。如微阵列数据所示,与正常精子发生对照相比,SCOS患者的hub基因表达水平显着降低,单细胞转录组数据,和临床样本水平。此外,通过miRNA-mRNA网络构建预测潜在的miRNA.这些hub基因和miRNA可用作可能与SCOS相关的潜在生物标志物。然而,目前尚未证明这些生物标志物的差异表达是SCOS的分子发病机制。我们的发现表明,这些生物标志物可以作为诊断目标的临床工具,并且可能从睾丸生殖细胞的角度对SCOS的精子发生产生一些影响。
    Sertoli cell-only syndrome (SCOS), a severe testicular spermatogenic failure, is characterized by total absence of male germ cells. To better expand the understanding of the potential molecular mechanisms of SCOS, we used microarray datasets from the Gene Expression Omnibus (GEO) and ArrayExpress databases to determine the differentially expressed genes (DEGs). In addition, functional enrichment analysis including the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) was performed. Protein-protein interaction (PPI) networks, modules, and miRNA-mRNA regulatory networks were constructed and analyzed and the validation of hub genes was performed. A total of 601 shared DEGs were identified, including 416 down-regulated and 185 up-regulated genes. The findings of the enrichment analysis indicated that the shared DEGs were mostly enriched in sexual reproduction, reproductive process, male gamete generation, immune response, and immunity-related pathways. In addition, six hub genes (CCNA2, CCNB2, TOP2A, CDC20, BUB1, and BUB1B) were selected from the PPI network by using the cytoHubba and MCODE plug-ins. The expression levels of the hub genes were significantly decreased in patients with SCOS compared to that in normal spermatogenesis controls as indicated by the microarray data, single-cell transcriptomic data, and clinical sample levels. Furthermore, the potential miRNAs were predicted via the miRNA-mRNA network construction. These hub genes and miRNAs can be used as potential biomarkers that may be related to SCOS. However, it has not been proven that the differential expression of these biomarkers is the molecular pathogenesis mechanisms of SCOS. Our findings suggest that these biomarkers can be serve as clinical tool for diagnosis targets and may have some impact on the spermatogenesis of SCOS from a testicular germ cell perspective.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:仅支持细胞综合征(SCOS)是非梗阻性无精子症最严重的病理类型。最近,已经确定了几个与SCOS相关的基因,包括FANCM,TEX14、NR5A1、NANOS2、PLK4、WNK3和FANCA,但不能完全解释SCOS的发病机制。本研究试图通过睾丸组织RNA测序来解释SCOS中的精子发生功能障碍,为SCOS的诊断和治疗提供新的靶点。
    方法:我们基于RNA测序分析了9例SCOS患者和3例梗阻性无精子症和正常精子发生患者的差异表达基因(DEGs)。我们使用ELISA和免疫组织化学进一步探索了鉴定的基因。
    结果:总计,在SCOS样品中表达了9406个DEGs(Log2|FC|≥1;调整后的P值<0.05),并鉴定了21个hub基因。发现了三个上调的核心基因,包括CASP4、CASP1和PLA2G4A。因此,我们推测CASP1和CASP4介导的睾丸细胞焦亡可能参与了SCOS的发生和发展。ELISA证实,SCOS患者睾丸中的CASP1和CASP4活性明显高于精子发生正常患者。免疫组织化学结果显示,CASP1和CASP4在正常生精组中主要表达于生精细胞核,Sertoli,和间质细胞。由于精原细胞和精母细胞的丢失,SCOS组的CASP1和CASP4主要在支持细胞和间质细胞核中表达。SCOS患者睾丸中CASP1和CASP4的表达水平明显高于正常生精患者。此外,SCOS患者睾丸中的焦亡相关蛋白GSDMD和GSDME也明显高于对照组.ELISA还显示炎症因子(IL-1β,IL-18,LDH,和ROS)在SCOS组中显著增加。
    结论:第一次,我们发现,在SCOS患者的睾丸中,细胞焦亡相关基因和关键标志物显著增加.我们还在SCOS中观察到许多炎症和氧化应激反应。因此,我们认为,CASP1和CASP4介导的睾丸细胞焦亡可能参与了SCOS的发生和发展。
    BACKGROUND: Sertoli cell-only syndrome (SCOS) is the most serious pathological type of non-obstructive azoospermia. Recently, several genes related to SCOS have been identified, including FANCM, TEX14, NR5A1, NANOS2, PLK4, WNK3, and FANCA, but they cannot fully explain the pathogenesis of SCOS. This study attempted to explain spermatogenesis dysfunction in SCOS through testicular tissue RNA sequencing and to provide new targets for SCOS diagnosis and therapy.
    METHODS: We analyzed differentially expressed genes (DEGs) based on RNA sequencing of nine patients with SCOS and three patients with obstructive azoospermia and normal spermatogenesis. We further explored the identified genes using ELISA and immunohistochemistry.
    RESULTS: In total, 9406 DEGs were expressed (Log2|FC|≥ 1; adjusted P value < 0.05) in SCOS samples, and 21 hub genes were identified. Three upregulated core genes were found, including CASP4, CASP1, and PLA2G4A. Thus, we hypothesized that testis cell pyroptosis mediated by CASP1 and CASP4 might be involved in SCOS occurrence and development. ELISA verified that CASP1 and CASP4 activities in the testes of patients with SCOS were significantly higher than those in patients with normal spermatogenesis. Immunohistochemical results showed that CASP1 and CASP4 in the normal spermatogenesis group were mainly expressed in the nuclei of spermatogenic, Sertoli, and interstitial cells. CASP1 and CASP4 in the SCOS group were mainly expressed in the nuclei of Sertoli and interstitial cells because of the loss of spermatogonia and spermatocytes. CASP1 and CASP4 expression levels in the testes of patients with SCOS were significantly higher than those in patients with normal spermatogenisis. Furthermore, the pyroptosis-related proteins GSDMD and GSDME in the testes of patients with SCOS were also significantly higher than those in control patients. ELISA also showed that inflammatory factors (IL-1 β, IL-18, LDH, and ROS) were significantly increased in the SCOS group.
    CONCLUSIONS: For the first time, we found that cell pyroptosis-related genes and key markers were significantly increased in the testes of patients with SCOS. We also observed many inflammatory and oxidative stress reactions in SCOS. Thus, we propose that testis cell pyroptosis mediated by CASP1 and CASP4 could participate in SCOS occurrence and development.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Meta-Analysis
    背景:目前还没有系统评价和荟萃分析来分析和总结挽救性微解剖睾丸精子提取(mTESE)中精子提取成功的预测因素。
    目的:我们旨在研究预测初次mTESE或常规睾丸精子提取(cTESE)失败的NOA患者mTESE挽救结果的因素。
    方法:我们在PubMed,WebofScience,EMBASE,和Cochrane图书馆提供的文献描述了在2022年6月之前发表的初始mTESE或cTESE未通过后接受抢救mTESE的NOA患者的特征。
    结果:本荟萃分析包括4项回顾性研究,对332例初次mTESE失败的NOA患者和3项回顾性研究,对177例cTESE失败的NOA患者。结果如下:在首次手术为mTESE的NOA患者中,年轻患者(SMD:-0.28,95%CI:-0.55至-0.01)和双侧睾丸体积(TV)较小的患者(SMD:-0.55,95%CI:-0.95至-0.15),较低水平的FSH(SMD:-0.86,95%CI:-1.18至-0.54)和LH(SMD:-0.68,95%CI:-1.16至-0.19),睾丸组织学类型为精子发生障碍(HS)(OR:3.52,95%CI:1.30至9.53)更有可能成功回收精子。而仅支持细胞综合征(SCOS)(OR:0.41,95%CI:0.24至0.73)的患者在挽救性mTESE中再次失败的可能性更大。此外,在初次cTESE失败后接受抢救mTESE的患者中,那些睾丸组织学类型的精子发生不足(HS)(OR:30.35,95%CI:8.27至111.34)更有可能成功,而那些成熟停滞(MA)(OR:0.39,95%CI:0.18至0.83)的患者很少受益。
    结论:我们发现年龄,电视,FSH,LH,HS,SCOS和MA是救助mTESE的有价值的预测因子,这将有助于男科医生的临床决策,并最大限度地减少对患者不必要的伤害。本文受版权保护。保留所有权利。
    BACKGROUND: There has been no systematic review and meta-analysis to analyze and summarize the predictive factors of successful sperm extraction in salvage microdissection testicular sperm extraction.
    OBJECTIVE: We aimed to investigate the factors predicting the result of salvage microdissection testicular sperm extraction in patients with non-obstructive azoospermia who failed the initial microdissection testicular sperm extraction or conventional testicular sperm extraction.
    METHODS: We conducted a systematic literature search in PubMed, Web of Science, EMBASE, and the Cochrane Library for literature that described the characteristics of patients with non-obstructive azoospermia who underwent salvage microdissection testicular sperm extraction after failing the initial microdissection testicular sperm extraction or conventional testicular sperm extraction published prior to June 2022.
    RESULTS: This meta-analysis included four retrospective studies with 332 patients with non-obstructive azoospermia who underwent a failed initial microdissection testicular sperm extraction and three retrospective studies with 177 non-obstructive azoospermia patients who underwent a failed conventional testicular sperm extraction. The results were as follows: among non-obstructive azoospermia patients whose first surgery was microdissection testicular sperm extraction, younger patients (standard mean difference: -0.28, 95% confidence interval [CI]: -0.55 to -0.01) and those with smaller bilateral testicular volume (standard mean difference: -0.55, 95% CI: -0.95 to -0.15), lower levels of follicle-stimulating hormone (standard mean difference: -0.86, 95% CI: -1.18 to -0.54) and luteinizing hormone (standard mean difference: -0.68, 95% CI: -1.16 to -0.19), and whose testicular histological type was hypospermatogenesis (odds ratio: 3.52, 95% CI: 1.30-9.53) were more likely to retrieve spermatozoa successfully, while patients with Sertoli-cell-only syndrome (odds ratio: 0.41, 95% CI: 0.24-0.73) were more likely to fail again in salvage microdissection testicular sperm extraction. Additionally, in patients who underwent salvage microdissection testicular sperm extraction after a failed initial conventional testicular sperm extraction, those with testicular histological type of hypospermatogenesis (odds ratio: 30.35, 95% CI: 8.27-111.34) were more likely to be successful, while those with maturation arrest (odds ratio: 0.39, 95% CI: 0.18-0.83) rarely benefited.
    CONCLUSIONS: We found that age, testicular volume, follicle-stimulating hormone, luteinizing hormone, hypospermatogenesis, Sertoli-cell-only syndrome, and maturation arrest were valuable predictors of salvage microdissection testicular sperm extraction, which will assist andrologists in clinical decision-making and minimize unnecessary injury to patients.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    仅支持细胞综合征(SCOS)是导致男性不育的一种睾丸病理衰竭,没有有效的治疗策略。可用于此条件。此外,其发展的分子机制仍然未知。我们基于从基因表达合成数据库获得的睾丸组织样品的四个数据集,将DExD/H-Box解旋酶58(DDX58)鉴定为SCOS中的关键基因。DDX58在SCOS睾丸支持细胞中显著上调。此外,DDX58的高表达与几种睾丸炎症因子的表达呈正相关,如IL-1β,IL-18和IL-6。有趣的是,在D-半乳糖(D-gal)刺激的TM4细胞损伤模型中可以诱导DDX58。而DDX58的沉默抑制了D-gal介导的p65表达,炎性细胞因子释放,增长停滞。机械上,我们发现DDX58作为RNA结合蛋白,通过促进mRNA稳定性来增强p65表达。此外,p65基因沉默降低了D-gal诱导的细胞中炎症细胞因子的表达和细胞生长的抑制。总之,我们的研究结果表明,DDX58通过稳定p65mRNA促进SCOS支持细胞的炎症反应和生长停滞.因此,DDX58/p65调节轴可能是SCOS的治疗靶点.
    Sertoli cell -only syndrome (SCOS) is a type of testicular pathological failure that causes male infertility and no effective treatment strategy, is available for this condition. Moreover, the molecular mechanism underlying its development remains unknown. We identified DExD/H-Box helicase 58 (DDX58) as a key gene in SCOS based on four datasets of testicular tissue samples obtained from the Gene Expression Synthesis database. DDX58 was significantly upregulated in SCOS testicular Sertoli cells. Moreover, high expression of DDX58 was positively correlated with the expression of several testicular inflammatory factors, such as IL -1β, IL-18, and IL-6. Interestingly, DDX58 could be induced in the D-galactose (D-gal)-stimulated TM4 cell injury model. Whereas silencing of DDX58 inhibited D-gal -mediated p65 expression, inflammatory cytokine release, and growth arrest. Mechanistically, we found that DDX58 acts as an RNA-binding protein, which enhances p65 expression by promoting mRNA stability. Furthermore, p65 gene silencing decreased the expression of inflammatory cytokines and inhibition of cell growth in D-gal-induced cells. In conclusion, our findings demonstrate that DDX58 promotes inflammatory responses and growth arrest in SCOS Sertoli cells by stabilizing p65 mRNA. Accordingly, the DDX58/p65 regulatory axis might be a therapeutic target for SCOS.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    男性不育可由精子发生的数量和/或质量异常引起,影响男性的身心健康。仅支持细胞综合征(SCOS)是男性不育的最严重的组织学表型,其特征是生殖细胞耗尽,仅支持细胞保留在生精小管中。大多数SCOS病例不能用已知的遗传原因来解释,包括染色体核型异常和Y染色体微缺失。随着测序技术的发展,近年来,关于筛查SCOS新遗传原因的研究越来越多。在散发性病例中对靶基因进行直接测序和在家族性病例中应用的全外显子组测序已经鉴定出与SCOS相关的几个基因。睾丸转录组的分析,蛋白质组,和SCOS患者的表观遗传学为SCOS的分子机制提供了解释。在这次审查中,我们基于具有SCO表型的小鼠模型讨论了种系发育缺陷与SCOS之间的可能关系。我们还总结了在探索SCOS遗传原因和机制方面的进展和挑战。了解SCOS的遗传因素可以更好地了解SCO和人类精子发生,对提高诊断水平也有现实意义,做出适当的医疗决定,和遗传咨询。对于治疗意义,SCOS研究,随着干细胞技术和基因治疗的成就,为SCOS患者开发新疗法以产生功能性精子奠定基础,给了他们孩子父亲的希望。
    Male infertility can be caused by quantitative and/or qualitative abnormalities in spermatogenesis, which affects men\'s physical and mental health. Sertoli cell-only syndrome (SCOS) is the most severe histological phenotype of male infertility characterized by the depletion of germ cells with only Sertoli cells remaining in the seminiferous tubules. Most SCOS cases cannot be explained by the already known genetic causes including karyotype abnormalities and microdeletions of the Y chromosome. With the development of sequencing technology, studies on screening new genetic causes for SCOS are growing in recent years. Directly sequencing of target genes in sporadic cases and whole-exome sequencing applied in familial cases have identified several genes associated with SCOS. Analyses of the testicular transcriptome, proteome, and epigenetics in SCOS patients provide explanations regarding the molecular mechanisms of SCOS. In this review, we discuss the possible relationship between defective germline development and SCOS based on mouse models with SCO phenotype. We also summarize the advances and challenges in the exploration of genetic causes and mechanisms of SCOS. Knowing the genetic factors of SCOS offers a better understanding of SCO and human spermatogenesis, and it also has practical significance for improving diagnosis, making appropriate medical decisions, and genetic counseling. For therapeutic implications, SCOS research, along with the achievements in stem cell technologies and gene therapy, build the foundation to develop novel therapies for SCOS patients to produce functional spermatozoa, giving them hope to father children.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:男性不育的最严重状况是完全的仅支持细胞综合征(SCOS),这是指睾丸中缺乏所有生精细胞。SCOS的遗传原因仍有待探索。我们旨在研究SCOS的遗传原因,并评估已鉴定的致病变异对人类男性生殖细胞的影响。
    方法:进行全外显子组测序以鉴定患有完整SCOS的男性的潜在致病变异,进行了Sanger测序以验证该男子及其父亲和兄弟的致病变异。通过将人诱导的多能干细胞(hiPSCs)体外分化为生殖细胞样细胞,研究了致病变体的致病机制。
    结果:PIWIL2中的纯合功能丧失(LoF)变体p.His244ArgfsTer31(c.731_732delAT)被鉴定为具有完整SCOS的男性的致病变体,在他的父亲和兄弟中证实了相同的杂合变异。该变体导致缺乏所有功能结构域的截短PIWIL2蛋白,并且在患者的睾丸中未检测到PIWIL2表达。患者和PIWIL2-/-hiPSCs可在体外分化为原始生殖细胞样细胞和精原干细胞样细胞(SSCLC),但SSCLCs的形成和维持严重受损。RNA-seq分析提示PIWIL2-/-组SSCLC诱导过程中Wnt信号通路失活,通过RT-qPCR在患者组中验证。Wnt信号通路抑制剂阻碍了正常hiPSCs分化过程中SSCLCs的形成和维持。
    结论:我们的研究揭示了PIWIL2在人类精原干细胞形成和维持中的关键作用。我们提供了临床和功能证据,证明PIWIL2中的LoF变异是SCOS的遗传原因,这支持了PIWIL2在遗传诊断中的潜在作用。此外,我们的结果强调了体外分化模型在功能验证实验中的适用性.
    BACKGROUND: The most serious condition of male infertility is complete Sertoli cell-only syndrome (SCOS), which refers to the lack of all spermatogenic cells in the testes. The genetic cause of SCOS remains to be explored. We aimed to investigate the genetic cause of SCOS and assess the effects of the identified causative variant on human male germ cells.
    METHODS: Whole-exome sequencing was performed to identify potentially pathogenic variants in a man with complete SCOS, and Sanger sequencing was performed to verify the causative variant in this man and his father and brother. The pathogenic mechanisms of the causative variant were investigated by in vitro differentiation of human-induced pluripotent stem cells (hiPSCs) into germ cell-like cells.
    RESULTS: The homozygous loss-of-function (LoF) variant p.His244ArgfsTer31 (c.731_732delAT) in PIWIL2 was identified as the causative variant in the man with complete SCOS, and the same variant in heterozygosis was confirmed in his father and brother. This variant resulted in a truncated PIWIL2 protein lacking all functional domains, and no PIWIL2 expression was detected in the patient\'s testes. The patient and PIWIL2-/- hiPSCs could be differentiated into primordial germ cell-like cells and spermatogonial stem cell-like cells (SSCLCs) in vitro, but the formation and maintenance of SSCLCs were severely impaired. RNA-seq analyses suggested the inactivation of the Wnt signaling pathway in the process of SSCLC induction in the PIWIL2-/- group, which was validated in the patient group by RT-qPCR. The Wnt signaling pathway inhibitor hindered the formation and maintenance of SSCLCs during the differentiation of normal hiPSCs.
    CONCLUSIONS: Our study revealed the pivotal role of PIWIL2 in the formation and maintenance of human spermatogonial stem cells. We provided clinical and functional evidence that the LoF variant in PIWIL2 is a genetic cause of SCOS, which supported the potential role of PIWIL2 in genetic diagnosis. Furthermore, our results highlighted the applicability of in vitro differentiation models to function validation experiments.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    仅支持细胞综合征(SCOS)是非梗阻性无精子症最严重和最常见的病理类型。尽管在该领域报道了少数研究,但迄今为止,SCOS的病因仍然未知。根据来自基因表达Omnibus的六个数据集的睾丸组织样本的基因表达,我们在SCOS和梗阻性无精子症(OA)睾丸组织样本之间检测到1441个差异表达基因(DEGs)。下调基因的GO术语和KEGG通路包括与细胞周期和繁殖相关的各种术语和通路,而上调基因的富集产生了许多与炎症相关的术语和途径。根据蛋白质-蛋白质相互作用(PPI)网络,最关键模块中的所有基因都属于下调的DEG,我们获得了九个hub基因,包括CCNB1,AURKA,CCNA2,BIRC5,TYMS,UBE2C,CDC20,TOP2A,OIP5在这些中枢基因中,在共识模块分析中获得的最重要的SCOS特定模块中也发现了6个.此外,大多数针对SCOS的模块没有共识。基于下调的基因,对上游调控网络中的转录因子(TFs)和激酶进行了预测。然后,我们比较了OA和SCOS样本中免疫细胞浸润水平的差异,发现SCOS中大多数免疫细胞的浸润程度明显高于OA样本.此外,CD56bright自然杀伤细胞与6个hub基因显著相关。SCOS中丰富的标志途径的上调途径明显多于下调途径。总的来说,我们探测到DEG,重要的模块,集线器基因,上游TFs和激酶,丰富的下游途径,和浸润的免疫细胞可能与SCOS的发病机理有关。这些发现为SCOS的发病机理提供了新的见解,并推动了其治疗方法的未来发展。
    Sertoli cell-only syndrome (SCOS) is the most severe and common pathological type of non-obstructive azoospermia. The etiology of SCOS remains largely unknown to date despite a handful of studies reported in this area. According to the gene expression of testicular tissue samples in six datasets from the Gene Expression Omnibus, we detected 1441 differentially expressed genes (DEGs) between SCOS and obstructive azoospermia (OA) testicular tissue samples. Enriched GO terms and KEGG pathways for the downregulated genes included various terms and pathways related to cell cycle and reproduction, while the enrichment for the upregulated genes yielded many inflammation-related terms and pathways. In accordance with the protein-protein interaction (PPI) network, all genes in the most critical module belonged to the downregulated DEGs, and we obtained nine hub genes, including CCNB1, AURKA, CCNA2, BIRC5, TYMS, UBE2C, CDC20, TOP2A, and OIP5. Among these hub genes, six were also found in the most significant SCOS-specific module obtained from consensus module analysis. In addition, most of SCOS-specific modules did not have a consensus counterpart. Based on the downregulated genes, transcription factors (TFs) and kinases within the upstream regulatory network were predicted. Then, we compared the difference in infiltrating levels of immune cells between OA and SCOS samples and found a significantly higher degree of infiltration for most immune cells in SCOS than OA samples. Moreover, CD56bright natural killer cell was significantly associated with six hub genes. Enriched hallmark pathways in SCOS had remarkably more upregulated pathways than the downregulated ones. Collectively, we detected DEGs, significant modules, hub genes, upstream TFs and kinases, enriched downstream pathways, and infiltrated immune cells that might be specifically implicated in the pathogenesis of SCOS. These findings provide new insights into the pathogenesis of SCOS and fuel future advances in its theranostics.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    大量男性不育是由无精子症引起的。然而,潜在的病因和分子基础在很大程度上仍然未知。通过单细胞(sc)RNA测序,我们分析了2例梗阻性无精子症(OA)和非梗阻性无精子症(NOA)患者的睾丸活检样本.我们在NOA样品中仅发现了体细胞,并探索了与生殖细胞相互作用丧失后支持细胞的转录变化。此外,我们观察到OA(输精管切除术后)患者和健康个体之间的生殖细胞群差异.我们在具有GSM3526588和GSE124263的两个数据集以进行详细分析的次要研究中证实了这一观察结果,其中鉴定了转录水平的调节机制。因此,这些发现为人类精子发生提供了有价值的信息,我们还确定了对生殖相关疾病的进一步研究有洞察力的信息。
    A substantial number of male infertility is caused by azoospermia. However, the underlying etiology and the molecular basis remain largely unknown. Through single-cell (sc)RNA sequencing, we had analyzed testis biopsy samples from two patients with obstructive azoospermia (OA) and nonobstructive azoospermia (NOA). We found only somatic cells in the NOA samples and explored the transcriptional changes in Sertoli cells in response to a loss of interactions with germ cells. Moreover, we observed a germ cell population discrepancy between an OA (postvasectomy) patient and a healthy individual. We confirmed this observation in a secondary study with two datasets at GSM3526588 and GSE124263 for detailed analysis wherein the regulatory mechanisms at the transcriptional level were identified. These findings thus provide valuable information on human spermatogenesis, and we also identified insightful information for further research on reproduction-related diseases.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号