Sertoli Cell-Only Syndrome

仅支持细胞综合征
  • 文章类型: Journal Article
    无精子症,精液中没有精子细胞,影响约15%的不育男性。仅支持细胞综合征(SCOS)是非梗阻性无精子症背景下最常见的病理病变,其特征是完全没有生发上皮,睾丸支持细胞仅存在于生精小管中。研究表明,成功的精子发生和男性生育力与精子的脂质组成之间存在相关性,精液,精浆或睾丸。这项研究的目的是发现Johnsen评分系统与SCOS患者睾丸冷冻切片中磷脂表达之间的相关性。MALDI成像质谱用于确定分子种类的空间分布,如磷脂。磷脂酰胆碱(PC),磷脂酰乙醇胺(PE)和鞘磷脂(SM)是哺乳动物细胞和睾丸中最丰富的磷脂。SM,质膜的结构成分,对精子发生和精子功能至关重要。疟原虫,是睾丸中独特的PC,具有很强的抗氧化性能。这项研究,使用成像质谱,展示了磷脂的局部分布,特别是SM,PC,首次使用SCOS在人睾丸样品中的疟原虫和PE。这项研究发现Johnsen评分系统与人睾丸组织中磷脂表达水平之间存在很强的关系。未来的发现可以在microTESE程序中实现常规诊断技术,以成功提取精子。
    Azoospermia, the absence of sperm cells in semen, affects around 15% of infertile males. Sertoli cell-only syndrome (SCOS) is the most common pathological lesion in the background of non-obstructive azoospermia and is characterised by the complete absence of germinal epithelium, with Sertoli cells exclusively present in the seminiferous tubules. Studies have shown a correlation between successful spermatogenesis and male fertility with lipid composition of spermatozoa, semen, seminal plasma or testis. The aim of this research was to discover the correlation between the Johnsen scoring system and phospholipid expressions in testicular cryosections of SCOS patients. MALDI imaging mass spectrometry is used to determine spatial distributions of molecular species, such as phospholipids. Phosphatidylcholines (PCs), phosphatidylethanolamines (PEs) and sphingomyelins (SMs) are the most abundant phospholipids in mammalian cells and testis. SMs, the structural components of plasma membranes, are crucial for spermatogenesis and sperm function. Plasmalogens, are unique PCs in testis with strong antioxidative properties. This study, using imaging mass spectrometry, demonstrates the local distribution of phospholipids, particularly SMs, PCs, plasmalogens and PEs in human testicular samples with SCOS for the first time. This study found a strong relationship between the Johnsen scoring system and phospholipid expression levels in human testicular tissues. Future findings could enable routine diagnostic techniques during microTESE procedures for successful sperm extraction.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Case Reports
    无精子症是男性不育的一种形式,其特征是射精中完全缺乏精子。仅支持细胞综合征(SCOS)是最严重的无精子症,在小管中没有发现生殖细胞。最近,据报道,FANCM基因变异是生精失败的新遗传原因。同时,已知FANCM变体与癌症易感性相关。我们对一名被诊断患有SCOS和健康父亲的男性患者进行了全外显子组测序。在患者中发现了两个FANCM基因的复合杂合错义突变,都是从父母那里继承的。不孕症评估后,患者被诊断为弥漫性星形细胞瘤。患者睾丸和肿瘤组织的免疫组织化学分析和适当的对照显示,第一次,不仅在星形细胞瘤中存在FANCM的细胞质而非核模式,而且在非有丝分裂神经元中也存在。在SCOS患者的睾丸组织中,细胞质抗FANCM染色强度似乎低于对照。我们的病例报告提出了一种新的可能性,即FANCM基因错义变异的不育携带者也容易发生癌症。
    Azoospermia is a form of male infertility characterized by a complete lack of spermatozoa in the ejaculate. Sertoli cell-only syndrome (SCOS) is the most severe form of azoospermia, where no germ cells are found in the tubules. Recently, FANCM gene variants were reported as novel genetic causes of spermatogenic failure. At the same time, FANCM variants are known to be associated with cancer predisposition. We performed whole-exome sequencing on a male patient diagnosed with SCOS and a healthy father. Two compound heterozygous missense mutations in the FANCM gene were found in the patient, both being inherited from his parents. After the infertility assessment, the patient was diagnosed with diffuse astrocytoma. Immunohistochemical analyses in the testicular and tumor tissues of the patient and adequate controls showed, for the first time, not only the existence of a cytoplasmic and not nuclear pattern of FANCM in astrocytoma but also in non-mitotic neurons. In the testicular tissue of the SCOS patient, cytoplasmic anti-FANCM staining intensity appeared lower than in the control. Our case report raises a novel possibility that the infertile carriers of FANCM gene missense variants could also be prone to cancer 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: 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: Case Reports
    睾丸间质细胞瘤(LCT)在成人中非常罕见。它仅占总睾丸肿瘤的1%。LCT可以产生类固醇激素,如雌激素,黄体酮,和睾丸激素。在生精小管中发现了支持细胞,它们是血睾丸屏障的一部分.仅支持细胞综合征(SCOS)也称为生殖细胞发育不全,其特征是无精子症,其中睾丸活检的生精小管仅衬有支持细胞。在正常T和LH的情况下,SCOS中预期的激素谱是FSH增加。LCT中的预期激素谱是增加的/正常的FSH和LH,具有增加的T或E2。一位患者出现在我们的诊所,他的右睾丸中有一个界限分明的肿块,并接受了根治性睾丸切除术。肿瘤标志物阴性。在精子图中检测到无精子症。T和E2正常,FSH,LH高。行根治性睾丸切除术。病理结果报告了LCT和SCOS的组合。在LCTs中经常遇到继发于高雄激素水平的无精子症病例。正如我们所介绍的那样,两种不同的睾丸病理可能同时出现,并产生意想不到的荷尔蒙图片。这种情况可能导致实验室掩盖临床事实。
    Leydig Cell Tumor (LCT) is very rare in adults. It constitutes only 1% of total testicular tumors. LCTs can produce steroid hormones such as estrogen, progesterone, and testosterone. Sertoli cells are found in seminiferous tubules, they are part of the blood-testis barrier. Sertoli Cells Only Syndrome (SCOS) also known as germ cell aplasia is characterized by azoospermia in which the seminiferous tubules of testicular biopsy are lined only with Sertoli cells. The expected hormone profile in SCOS is increased FSH with normal T and LH. The expected hormone profile in LCT is increased/normal FSH and LH with increased T or E2. A patient presented to our clinic with a well-circumscribed mass in his right testicle and underwent radical orchiectomy. Tumor markers were negative. Azoospermia was detected in the spermiogram. T and E2 were normal, FSH, and LH were high. Right radical orchiectomy was performed. A combination of LCT and SCOS were reported in pathology results. Azoospermia cases secondary to high androgen levels are frequently encountered in LCTs. As in the case we have presented, two different testicular pathologies may present at the same time and create an unexpected hormonal picture. Such situations can cause the laboratory to mask the clinical truth.
    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
    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: 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: Case Reports
    睾丸癌占男性全部癌症的1-1.5%,性索间质肿瘤包括5%的睾丸癌。这项研究旨在报告同一患者同时出现的仅支持细胞综合征和睾丸间质细胞肿瘤。一名32岁男子,有3年的原发性不孕症病史。体格检查显示正常的第二性征。两次连续的精液分析显示无精子症。阴囊超声扫描显示28×27毫米的低回声和高血管右睾丸肿块。进行右睾丸根治性切除术并同时进行左睾丸活检。组织病理学检查显示仅支持细胞综合征和Leydig细胞肿瘤伴有局灶性Leydig细胞增生。在文献中很少提到在治疗Leydig细胞肿瘤后逆转生育能力。一项研究表明,原发性不育男性在经过4个月的管理后,生育能力得以恢复。然而,由于SCOS导致的非阻塞性无精子症的不育男性只能通过睾丸精子提取技术生一个孩子。尽管Leydig细胞瘤很少发生,在患有无精子症的不育男性中,可以看到它与仅支持细胞综合征相关。临床检查和影像学检查对这些患者很重要,因为其中睾丸肿块的可能性很高。
    Testicular cancers comprise 1-1.5% of entire cancers in men, and sex cord-stromal tumors include 5% of testicular cancers. This study aims to report a simultaneous Sertoli cell-only syndrome and Leydig cell tumor in the same patient. A 32-year-old man presented with a history of primary infertility for 3 years. Physical examination revealed normal secondary sexual characteristics. Two successive seminal fluid analyses revealed azoospermia. A scrotal ultrasound scan showed a 28 × 27 mm hypoechoic and hypervascular right testicular mass. Right radical orchiectomy and simultaneous left testicular biopsy were conducted. The histopathological examination revealed Sertoli cell-only syndrome and Leydig cell tumor with focal Leydig cell hyperplasia. Reversing fertility following the management of Leydig cell tumor is rarely mentioned in the literature. A study revealed that fertility recovered following 4 months of management in a primary infertile male. However, infertile men with nonobstructive azoospermia due to SCOS can only have a child by testicular sperm extraction technique. Despite the rare occurrence of Leydig cell tumor, it could be seen in association with Sertoli cell-only syndrome in infertile men with azoospermia. Clinical examination and imaging studies are important in these patients as the possibility of having a testicular mass is high among them.
    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
    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)

公众号